Sie sind auf Seite 1von 33

R E V I E W

The Osteocyte: An Endocrine Cell . . . and More

Sarah L. Dallas, Matthew Prideaux, and Lynda F. Bonewald


Department of Oral and Craniofacial Sciences, School of Dentistry, University of Missouri-Kansas City, Kansas City,
Missouri 64108

Few investigators think of bone as an endocrine gland, even after the discovery that osteocytes produce cir-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


culating fibroblast growth factor 23 that targets the kidney and potentially other organs. In fact, until the last
few years, osteocytes were perceived by many as passive, metabolically inactive cells. However, exciting recent
discoveries have shown that osteocytes encased within mineralized bone matrix are actually multifunctional
cells with many key regulatory roles in bone and mineral homeostasis. In addition to serving as endocrine cells
and regulators of phosphate homeostasis, these cells control bone remodeling through regulation of both
osteoclasts and osteoblasts, are mechanosensory cells that coordinate adaptive responses of the skeleton to
mechanical loading, and also serve as a manager of the bone’s reservoir of calcium. Osteocytes must survive for
decades within the bone matrix, making them one of the longest lived cells in the body. Viability and survival
are therefore extremely important to ensure optimal function of the osteocyte network. As we continue to
search for new therapeutics, in addition to the osteoclast and the osteoblast, the osteocyte should be considered
in new strategies to prevent and treat bone disease. (Endocrine Reviews 34: 658 – 690, 2013)

I. Introduction location embedded within mineralized bone matrix rather


II. Osteocyte Differentiation and Embedding than by their function. This is because until the past decade
III. Morphology of Osteocytes, Their Dendrites, and La- or so there has been a lack of clear understanding about the
cunocanalicular System
properties of these cells and their important functions in
IV. Osteocyte Selective Genes/Proteins and Their Potential
Functions the skeleton. This perception persisted despite the fact that
V. Tools for Studying Osteocytes osteocytes make up over 95% of the bone cells in the adult
VI. Osteocyte Mechanosensation and Transduction skeleton, with this ratio increasing with age and with the
A. How osteocytes sense loading size of the bone.
B. How loading affects osteocyte signaling Osteocytes reside in lacunae within the mineralized
VII. Osteocytes as Orchestrators of Bone Formation and bone matrix and send their dendritic processes (ranging
Resorption
from 40 –100 per cell [1]) through tiny tunnels called
VIII. Osteocyte Life, Death, and in Between
IX. Osteocytic Perilacunar Remodeling: An Old Concept canaliculi to form the osteocyte lacunocanalicular net-
Rediscovered work (Figures 1 and 2), which connects to cells on the bone
X. The Osteocyte as an Endocrine Cell surface and to the vasculature. A fluid, termed canalicular
XI. Crosstalk Between Osteocytes and Muscle Cells or bone fluid, that is still not well characterized travels
XII. Role of Osteocytes in Disease through the lacunocanalicular space and bathes the os-
XIII. Summary and Perspective teocyte, thereby providing oxygen and nutrients to main-
tain the viability of the cell in this enclosed environment.
Early bone histologists hypothesized various functions
I. Introduction for osteocytes, but lacked the tools to test their hypotheses.
f the major cell types in bone, osteoblasts and oste- Today, with the use of transgenic technologies combined
O oclasts have been defined by their respective func-
tions of bone formation and bone resorption, but osteo-
Abbreviations: ASARM, acidic serine aspartate-rich MEPE-associated motif; DKK1, Dick-
kopf1-related protein 1; DMP1, Dentin matrix protein 1; ER-␣, estrogen receptor-␣; FAK,
focal adhesion kinase; FGF, fibroblast growth factor; FGFR, FGF receptor; GFP, green
cytes were defined primarily by their morphology and fluorescent protein; kb, kilobase; LRP, low-density lipoprotein receptor-related protein;
MEPE, matrix extracellular phosphoglycoprotein; NO, nitric oxide; NOS, NO synthase;
ISSN Print 0163-769X ISSN Online 1945-7189 1,25(OH)2D, 1,25-dihydroxyvitamin D; OPG, osteoprotegerin; PC1, polycystin 1; PGE2,
Printed in U.S.A. prostaglandin E2; PHEX, phosphate-regulating gene with homologies to endopeptidases
Copyright © 2013 by The Endocrine Society on the X chromosome; PKA, protein kinase A; PKD1, polycystic kidney disease 1; PTH1R,
Received May 2, 2012. Accepted April 16, 2013. PTH receptor type 1; RANK, receptor activator of nuclear factor ␬-B; RANKL, RANK ligand;
First Published Online April 26, 2013 SFRP1, secreted frizzled-related protein 1; TRAP, tartrate-resistant acid phosphatase.

658 edrv.endojournals.org Endocrine Reviews, October 2013, 34(5):658 – 690 doi: 10.1210/er.2012-1026
doi: 10.1210/er.2012-1026 edrv.endojournals.org 659

Figure 1. teoblast and osteoclast function. Osteocytes


also act as mechanosensors to control adaptive
responses to mechanical loading of the skele-
ton, and they may be a key target cell for the
actions of PTH in bone. The osteocyte there-
fore appears to integrate hormonal and me-
chanical signals in the regulation of bone mass.
Because osteocytes reside in an enclosed en-
vironment for extended periods of time, which
can last up to decades, the viability of these cells
becomes critical for their function. Not only

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


does the viable osteocyte regulate bone homeo-
stasis, but the dying or apoptotic osteocyte also
may play key regulatory roles in sending sig-
nals to initiate bone remodeling, particularly
when there is a need for bone repair. Au-
tophagy (the process of controlled “self diges-
tion” of the cell contents) also appears to be
necessary to sustain the cell within the enclosed
environment of the mineralized matrix.
Another exciting and unexpected recent dis-
covery is that osteocytes may function in an
endocrine manner to regulate phosphate ho-
meostasis through secretion into the circula-
tion of fibroblast growth factor 23 (FGF23).
When one thinks of an endocrine organ, tissues
such as the pituitary or adrenal glands come to
mind, but one would not normally ascribe this
function to bone. However, kidney, liver, and
heart have all been shown to have endocrine
Figure 1. The osteocyte. Schematic representation of an embedded osteocyte
located within its lacuna, illustrating its dendritic processes passing through the bone
functions, and now it appears that bone (and
matrix (gray shading) within narrow tunnels termed canaliculi. The osteocyte’s specifically the osteocyte network) can be
dendritic processes interconnect with other osteocytes as well as surface osteoblasts. added to this category. Criteria for designating
Note that some osteocyte processes may extend beyond the osteoblast layer to
an organ as an endocrine gland are that it must
potentially interact with cells in the marrow and that osteocyte dendrites are also in
intimate contact with the vasculature. The composition of the perilacunar matrix form a system that directly secretes hormones
immediately adjacent to the osteocyte (mauve shading) is different from that of the into the bloodstream to affect distant target
rest of the bone matrix (gray shading), which may influence the magnitude of organs and cells. Endocrine organs must also
mechanical strains perceived by the osteocyte.
be highly vascularized. Bone meets these crite-
ria because it is a highly vascular organ and the
with molecular and cell biology and state-of-the-art in- osteocyte lacunocanalicular network is in intimate con-
strumentation, dramatic advances have been made in nection with this vascular supply (Figures 1 and 2). The
identifying and understanding the functions of these pre- kidney produces factors such as erythropoietin, and the
viously overlooked bone cells. It is now clear that, far from liver produces IGF, both of which can be considered hor-
being a passive cell as it has been historically described in mone-like. Similarly, the circulating factors produced by
the literature, the osteocyte is a key cell that is indispens- osteocytes, such as FGF23 and sclerostin, should also be
able for the normal function of the skeleton. Although considered in this category.
osteocytes most likely do not play a major role in embry- In summary, there has been an explosion in research on
onic bone development and may only play a secondary the properties and functions of osteocytes over the past
role in growth and development postnatally, they appear few years, leading to many exciting discoveries and the
to play a critical role in bone homeostasis in the adult assignment of new functions to this intriguing cell. This
skeleton. Thus, the osteocyte plays multifunctional roles review aims to summarize the current state of knowledge
in orchestrating bone remodeling by regulating both os- of the osteocyte and to review its many functions, includ-
660 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

Figure 2.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


Figure 2. Osteocyte morphology and interaction with the vasculature. A, Confocal fluorescence image of osteocytes in the cortex of an adult
mouse femur stained with Alexa Fluor 488 phalloidin, which stains the actin cytoskeleton of the cell. Note the highly dendritic morphology of the
osteocyte with many actin-positive dendrites extending from the cell in all directions. Note also the extensive dendritic interconnections between
adjacent osteocytes. B, Confocal fluorescence image of the osteocyte lacunocanalicular system in mouse cortical bone as revealed by the tracer
dye, procion red. This dye is dispersed throughout the lacunocanalicular system within only 5 minutes of iv injection, demonstrating the intimate
connection of the canalicular fluid with the circulation (as modified from Ref. 231). C, Scanning electron micrograph of an acid-etched resin-casted
mouse cortical bone specimen, revealing the 3-dimensional organization of the osteocyte lacunocanalicular system and its interaction with a blood
vessel (BV). Note the 2 osteocyte lacunae (OCY) and extensive canaliculi, which are intimately connected to the surface of the blood vessel. Also
note the extensive, interconnected canaliculi throughout the bone matrix.

ing its novel function as an endocrine cell. The review will ulation of osteoblasts on the bone surface slows down
also discuss the implications for treatment of bone diseases their matrix production relative to adjacent cells. These
and future directions for research of this fascinating and osteoblasts then become “buried alive” under the matrix
unique cell. produced by neighboring osteoblasts (2, 8, 11). However,
research from two different groups suggests that the em-
bedding mechanism may actually be an active and invasive
II. Osteocyte Differentiation and Embedding process in which matrix degradation is required for the
formation of the osteocyte lacuna and canaliculi (12, 13).
It is well accepted that osteocytes are derived from osteo-
During the transition process, one would expect the
blasts, which in turn are derived from osteogenic precur-
differentiating osteocytes to share many markers with
sors residing in the bone marrow. The mature osteocyte
their osteoblast precursors, but also to express unique
represents a terminally differentiated stage of the osteo-
marker genes that play key regulatory roles in altering
blast lineage. However, the precise mechanisms by which
their morphology and function. Until recently, character-
an osteoblast becomes embedded in bone matrix to begin
a new life as an osteocyte and the genetic and molecular izing the molecular and genetic changes that occur during
mechanisms that regulate the differentiation and matura- osteoblast to osteocyte differentiation was challenging
tion of the osteocyte are still not fully understood (re- due to the lack of appropriate osteocyte cell lines and dif-
viewed in Refs. 2– 4). Once an osteoblast stops actively ficulties in isolating sufficient numbers of osteocytes from
forming new bone matrix, it is thought to have one of three tissue sources. However, within the past two decades,
potential fates. It can become a quiescent cell on the bone more and more markers of the osteocyte have been iden-
surface, known as a lining cell (5), it can undergo pro- tified (reviewed in Refs. 3, 14, and 15). Furthermore, the
grammed cell death via the process of apoptosis (5, 6), or generation of transgenic mouse lines in which green flu-
it can become embedded in its own osteoid and differen- orescent protein (GFP) variants are selectively expressed in
tiate into an osteocyte. Several different transitional stages osteoblasts and osteocytes (16 –18) and the use of pro-
between osteoblasts and osteocytes have been reported in teomic and gene array technologies (19 –21) have enabled
the literature (7–10), based mainly on morphological ob- comparisons of gene and protein expression profiles be-
servations (reviewed in Ref. 2). Figure 3 shows a schematic tween osteoblasts and osteocytes, which has provided
representation of these transitional stages, compiled from clues to help identify the key mediators of osteocyte dif-
these various articles. ferentiation and function. The changes in gene expression
The embedding of an osteoblast to become an osteocyte that represent a signature for transition toward an osteo-
has been viewed as a passive process in which a subpop- cytic phenotype include down-regulation of type I colla-
doi: 10.1210/er.2012-1026 edrv.endojournals.org 661

Figure 3.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020

Figure 3. Osteocyte differentiation. A, Schematic diagram depicting the transitional stages that occur as osteoblasts differentiate into mature osteocytes. During
this process, the volume of the cell body and the number of cell organelles decreases. 1 ⫽ preosteoblast; 2 ⫽ osteoblast; 3 ⫽ embedding osteoblast; 4 ⫽ osteoid
osteocyte; 5 ⫽ mineralizing osteocyte; 6 and 7 ⫽ mature osteocytes. B, Tetrachrome-stained section of an adult mouse tibia illustrating several of the osteoblast-
osteocyte transitional stages depicted schematically in panel A (bar ⫽ 25 ␮m). C, Table illustrating the relative temporal expression of various osteogenic markers
during the transition from osteoblast to osteocyte as depicted in panels A and B. Runt-related transcription factor 2 (RUNX2) directs early osteoblast
differentiation and is expressed in both preosteoblasts and osteoblasts. Osteocalcin (OCN) is expressed by mature osteoblasts and early osteocytes. E11 is the
earliest osteocyte marker to be expressed during differentiation but is not found in mature osteocytes in vivo. DMP1, CapG, and MEPE expression is observed in
mineralizing and mature osteocytes, whereas sclerostin expression is confined to mature osteocytes. ORP150 is also only found in mature osteocytes within the
hypoxic environment of the mineralized bone matrix.
662 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

gen and alkaline phosphatase and up-regulation of genes produced by its neighbors. Marotti’s theory is based entirely
including dentin matrix protein 1 (DMP1), E11/GP38/ on morphological observations, but nevertheless, it is an at-
podoplanin, fibroblast growth factor 23 (FGF23), matrix tractive hypothesis that potentially explains the ordered
extracellular phosphoglycoprotein (MEPE), and phos- spacing of osteocytes in 3 dimensions. Our own studies using
phate-regulating gene with homologies to endopeptidases live cell imaging techniques have suggested that the osteo-
on the X chromosome (PHEX) (2, 22–25). Induction of cyte-embedding process is highly dynamic and that the em-
SOST, which encodes the protein sclerostin (26), indicates bedding cells repeatedly extend and retract their dendrites,
differentiation to a mature osteocyte phenotype. Com- making transient connections with already embedded osteo-
pared to osteoblasts, osteocytes are also enriched in pro- cytes (4, 40, 41). This “sampling” of the local environment
teins associated with resistance to hypoxia, such as through the use of “exploratory dendrites” may allow the
ORP150 (20), and proteins important in cytoskeletal osteocyte to position itself with an appropriate spacing from

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


function, including destrin, CapG, Cdc42, and E11 (19 – other embedded osteocytes to maintain the ordered 3-dimen-
21, 27, 28). Osteocytes also show high expression of sional spacing of the osteocyte network.
CD44 (29) and neuropeptide Y (30). This diverse set of Although the molecular and genetic mechanisms that
genes has functions ranging from regulation of mineral- regulate osteocyte differentiation are slowly being unrav-
ization (DMP1, SOST) to phosphate homeostasis (PHEX, eled, there remain many unanswered questions. These in-
MEPE, FGF23) and cytoskeletal arrangement and/or clude the question of whether every osteoblast has an
dendrite formation (E11, CD44, CapG, Cdc42, and equal chance of becoming an osteocyte or whether there
destrin). The specific functions of these osteocyte marker are specific subpopulations of osteoblasts with predefined
proteins are discussed in more detail in Section IV. fates. Aubin and colleagues (42, 43) have reported exten-
Although considerable progress has been made in gen- sively on the heterogeneity of surface osteoblasts in terms
erating a list of osteocyte marker genes, it remains unclear of their gene and protein expression profiles. It therefore
which of these are actually the key mediators that control seems plausible that there are specific subpopulations of
osteocyte differentiation. Notable among them is the osteoblasts on the bone surface that are already committed
membrane-bound protein E11, which is expressed in early to becoming an osteocyte. Another unresolved issue is
osteocytes (22, 31) and has been shown to play a role in the whether the decision for an osteoblast to differentiate into
formation of dendrites in osteocytes (32), a critical step in an osteocyte is a cell autonomous response or one that is
their differentiation. DMP1 may also play a role in osteo- controlled through signaling to surface osteoblasts from
cyte differentiation because deletion of this gene in mice osteocytes that are already embedded. Imai et al (44) pro-
leads to a delay in osteocyte maturation (33). Sclerostin posed that osteocytes can recruit osteoblasts and stimulate
has also recently been implicated as a regulator of the their differentiation by expressing osteoblast stimulating
differentiation from late osteoblast to preosteocyte factor-1 (OSF-1; also known as heparin-binding growth
through its regulation of PHEX and MEPE (34). Interest- associated molecule, HB-GAM). They postulated that
ingly, oxygen tension may also regulate the differentiation these osteoblasts then further differentiate into osteocytes,
of osteoblasts into osteocytes (35), suggesting that some of thereby providing a new source of OSF-1 for the next cycle
the genes associated with resistance to hypoxia may play of osteoblast recruitment.
a role in osteocyte differentiation. In addition, mineral- The life span of the osteocyte is most likely determined
ization of the extracellular matrix surrounding the osteo- by rates of bone turnover, the process by which osteoclasts
cyte has also been shown to induce osteocyte maturation, resorb bone and osteoblasts replace the resorbed bone.
both in vitro (36) and in vivo (37, 38). Osteocytes may have a half-life of decades if they are lo-
The embedding of osteocytes does not seem to be a cated within a bone that has a slow turnover rate. An
random process. When viewed 3-dimensionally, osteo- important question is whether the differentiation into an
cytes appear to be regularly spaced within the bone matrix, osteocyte is a “one-way trip,” ie, is it an irreversible pro-
with approximately equal distances between adjacent os- cess or do osteocytes have the ability to dedifferentiate
teocytes. It remains unclear what dictates this highly or- back to an osteoblast or potentially give rise to other lin-
dered 3-dimensional organization of the osteocyte net- eages, such as adipocytes and chondrocytes? The fate of
work. One interesting hypothesis comes from the work of osteocytes released by osteoclastic resorption is presently
Marotti (39), who has proposed that newly formed os- unknown. Some of the osteocytes that are only partially
teocytes secrete an osteoblast inhibitory signal once their exposed by resorptive activity may be re-embedded when
dendrites have reached their maximal length. This inhibits new bone is formed after resorption (45). Resorption
osteoid production of the nearest, most intimately con- could also potentially liberate the osteocyte, but there is
nected osteoblast, which then becomes buried in matrix little clear evidence that osteocytes dedifferentiate back
doi: 10.1210/er.2012-1026 edrv.endojournals.org 663

into the osteoblastic state (46). It seems most likely that the mineralization of the osteoid, there are also changes to
osteocytes exposed during bone resorption die by apopto- osteocyte ultrastructure, such as a reduction in the endo-
sis and/or become phagocytosed by osteoclasts, as has plasmic reticulum and Golgi apparatus (55). In the past,
been described in several reports (47– 49). With the avail- this has been interpreted to imply reduced protein syn-
ability of transgenic mouse models in which lineage-tar- thesis and metabolic activity of the osteocyte, leading to
geted GFP variants are expressed in osteoblasts and os- the misconception that osteocytes are passive, inactive
teocytes (16 –18) and the availability of transgenic tools cells and are of little importance during bone growth
for gene deletion and manipulation of gene expression in and development. However, it is now clear, as empha-
osteocytes (50, 51), it seems likely that the answers to some sized throughout this review, that osteocytes are highly
of these unresolved questions will soon be known and that active cells that play major roles in many key physio-
we will gain deeper and deeper insight into the mecha- logical processes both within and beyond the bone

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


nisms controlling osteocyte differentiation. microenvironment.
The transition from a plump polygonal osteoblast to a
stellate, highly dendritic osteocyte represents a profound
III. Morphology of Osteocytes, Their Dendrites, change, which presumably requires extensive reorganization
and Lacunocanalicular System of cytoskeletal proteins. The characteristic polarization of
the osteoblast is lost (56), and this is accompanied by striking
Osteocytes are one of an elite group of cells within the changes in the expression and localization of actin-binding
body, which also includes cementocytes and hypertrophic proteins, such as fimbrin, filamin, and ␣-actinin, as shown by
chondrocytes, that are completely encased within a min- Kamioka et al (57). These authors proposed that the orga-
eralized extracellular matrix. This presents significant nized expression of tubulin, vimentin, and actin in the cell
challenges with regard to communication with other cells bodies and dendrites of osteocytes is important to establish
and access to sufficient nutrients and oxygen. However, and maintain their unique dendritic morphology (58).
the osteocyte and its lacunocanalicular system are exqui- At first glance, the osteocyte may appear to be isolated
sitely adapted for this role and have developed a unique from neighboring osteocytes in the mineralized matrix and
physiology to cope with the challenges of being “impris- from osteoblasts on the bone surface. However, these cells
oned” within a mineralized matrix. show a high degree of interconnectivity. The cell body is
Unlike the molecular regulatory mechanisms, the mor- enclosed within a lacuna of 15–20 ␮m in diameter, and the
phological changes that occur during the transition from cell processes pass through the bone matrix through nar-
an osteoblast to an osteocyte have been well characterized row canals called canaliculi, approximately 250 –300 nm
(2, 10, 11, 52, 53). During differentiation, osteoblast mor- in diameter (Figures 1 and 2). Together, the osteocyte la-
phology changes from a polygonal shape to a more stellate cunae and canaliculi are referred to as the lacunocanalicu-
shape. One of the first changes to take place in the em- lar system. Numerous dendritic processes connect osteo-
bedding cell is the formation and elongation of numerous cytes to other osteocytes and allow osteocytes near the
cellular projections, or dendritic processes, which are ex- bone surface to be connected to osteoblasts and bone-
tended in a polarized manner toward the mineralizing lining cells. This characteristic morphology allows for the
front. This is followed by dendrites extending toward the passage of nutrients and biochemical signals from one os-
vascular space or bone surface. These long, slender cyto- teocyte to another, and through these interconnections,
plasmic processes radiate in all directions around the the embedded osteocytes form a functional network (re-
cell body of the osteocyte, with the highest density per- viewed in Refs. 15, 59, and 60). This interconnectivity of
pendicular to the bone surface (Figure 2, A and B). Once the osteocytes facilitates communication and helps main-
embedded, the osteocyte maintains this polarity in the di- tain cell viability and is therefore essential for the normal
rectionality of its dendrites and the directionality of min- functioning of the osteocyte. Disruption of the network
eralization such that mineral deposition occurs on 1 side can have negative consequences for bone health. To enable
of the embedding cell rather than equally all around it osteocytes to communicate with one another, gap junc-
(Figure 3B). We have previously proposed that the osteoid tions are present on the tips of the dendrites connecting
osteocyte (ie, the cell that is in the process of transitioning them (61). These are transmembrane channels that con-
from osteoblast to osteocyte), is the cell that is primarily nect the cytoplasm of adjacent cells and allow the cells to
responsible for mineralization as opposed to osteoblasts exchange molecules of less than 1 kDa (62).
on the bone surface (40, 54). Intriguingly, the work of Kamioka et al (63) suggests
As the osteocyte embeds and further differentiates, that some osteocyte cell processes extend beyond the os-
there is a reduction in cell volume of up to 70% (10). Upon teoblast layer, establishing direct contact between the os-
664 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

teocyte network and the marrow space. Through these osteocyte’s relationship with its lacunocanalicular system.
dendrites there is the potential for direct signaling between Nevertheless, by developing such an intricate network of
osteocytes and cells in the bone marrow compartment. lacunae and canaliculi and by adopting its unique mor-
Our studies using live cell imaging approaches suggest phology with extensive dendritic connections, the osteo-
that osteocytes, particularly those that have recently cyte has been able to overcome the challenges of living in
embedded, can extend and retract their dendritic pro- the hostile environment of the mineralized bone matrix.
cesses, not only toward other osteocytes and osteo-
blasts, but also into marrow spaces (64). This suggests
that osteocytes have the potential to make and break IV. Osteocyte Selective Genes/Proteins and
contact with other cell types. Their Potential Functions
The morphology of the lacunocanalicular system can

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


be demonstrated using techniques such as scanning elec- Determining genetic markers for osteocytes has proven to
tron microscopy (EM) of plastic-embedded, acid-etched be difficult due to the relatively low number of known
bone samples (65) or injection of mice with dyes such as osteocyte-specific genes. The recent characterization of
the small molecular weight dye, procion red (65, 66) (Fig- proteins such as E11, sclerostin, DMP1, and MEPE as
ure 2, B and C). These techniques reveal the intimate con- osteocyte markers has, however, enabled easier identifi-
nection of the osteocyte lacunocanalicular system with the cation of osteocytes as well as providing insight into the
vasculature and illustrate an important property of the functions of these cells.
lacunocanalicular system, namely that the canalicular The earliest marker known to be expressed by the dif-
space between the osteocyte dendrite and the canalicular ferentiating osteocyte is E11 (22, 31). In the adult mouse,
wall is filled with circulating fluid, canalicular fluid, that this marker is specifically expressed in osteocytes but not
carries solutes to and from the osteocyte via the circula- osteoblasts (32). Peak expression is observed in the young
tion. Dye-injection studies have shown permeation of the osteocytes, in comparison with the more mature cells
osteocyte canalicular space from the bloodstream within deeper in the mineralized bone matrix. Regulation of E11
only a few minutes after injection (66). Tracer studies have is poorly understood; however, the transcription factors
shown that molecules of less than 40 kDa can readily reach SP1/3 (77) and AP1 (78) are known to modulate expres-
the lacunocanalicular system from the circulation but that sion, and E11 is susceptible to post-translational degra-
molecules as large as 440 kDa cannot (67). The upper dation by the calpain family of proteases (79). E11 is
cutoff appears to be less than 70 kDa, and a filter of some known to bind to CD44 (80) and ezrin-radixin-moesin
sort appears to exist between the vascular system and the complexes; therefore, these molecules may function to-
osteocyte lacunocanalicular system (68). As discussed in gether to regulate the formation of dendritic processes in
Section VI, mechanical loading of bone results in the flow osteocytes, a critical step in their differentiation. Overex-
of canalicular fluid around the osteocyte and its dendrites, pression of E11 in vitro has been shown to induce dendrite
which may provide a key stimulus for mechanotransduc- formation in an osteoblast-like cell line (81), and similar
tion (reviewed in Refs. 14, 60, 68, and 69). It is thought cytoskeletal rearrangements were observed in keratino-
that the lacunar and canalicular spaces are not completely cytes, with relocalization of ezrin to cell surface projec-
open but are filled with proteoglycans that constitute the tions (82). Furthermore, small interfering RNA-mediated
glycocalyx of the cell (reviewed in Refs. 59 and 70). These knockdown of E11 prevented the fluid flow-induced elon-
may function as a molecular sieve that allows the rapid gation of the cell processes of MLO-Y4 osteocyte-like cells
passage of smaller molecular mass molecules (eg, ⬍70 (32). In addition to regulating dendrite elongation, bind-
kDa) but much slower passage of larger molecules (66). ing of E11 to the ezrin-radixin-moesin complexes activates
Interestingly, the morphology of the lacunocanalicular the small GTPase RhoA, and this was shown to result in
system may not be permanent. Several studies suggest that increased motility of an epithelial cell line (83). In vivo,
osteocytes can modify their microenvironment by depos- E11 expression was found to be increased in osteocytes
iting and resorbing bone around their lacunae (71–75). after mechanical loading (32). Deletion of E11 in mice did
Furthermore, there is evidence that the number of canalic- not significantly affect embryonic bone development;
uli increases with age (13, 76). This suggests either that the however, the effects of E11 on postnatal bone formation
new bone formed in older animals produces osteocytes are unknown because these mice die soon after birth due
with more canaliculi or that existing osteocytes can gen- to respiratory failure (84).
erate new dendrites after they have embedded, presumably Several members of the SIBLING (small, integrin-bind-
through a matrix degradative process (13). This adds an ing ligand, N-linked glycoprotein) family of proteins are
interesting changing dimension to considerations of the also highly expressed by osteocytes and are known to have
doi: 10.1210/er.2012-1026 edrv.endojournals.org 665

important functions within the bone environment. DMP1 Mutations in Phex, such as in the Hyp mouse, or loss of
expression has been described in hypertrophic chondro- PHEX expression leads to increased levels of FGF23
cytes and osteoblasts during embryonic bone formation through an unknown mechanism and this leads to subse-
(85); however, its expression is more restricted to osteo- quent hypophosphatemia. Interestingly, FGF23 is not
cytes during postnatal development (23). One of the main normally expressed at high levels in osteocytes in the
functions of DMP1 is thought to be in the regulation of healthy state, but its expression in osteocytes is dramati-
mineralization, consistent with its pattern of expression cally up-regulated in both DMP1- and PHEX-associated
within the bone (87). Further evidence for this is provided hypophosphatemic rickets (92). Moreover, osteocytes ap-
by Dmp1-null mice, which show hypophosphatemia and pear to be the main source of the elevated circulating levels
highly elevated levels of FGF23, leading to a severe im- of FGF23 seen in these mouse models, giving support to
pairment in mineralization and resulting in an osteoma- the notion that they act as endocrine cells.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


lacic phenotype (65). In contrast to loss of function, the FGF23 expression can also be regulated by another SIB-
gain of function effects of DMP1 in vivo are less clear. Lu LING protein, MEPE, which is predominantly expressed
et al (33) showed no effect of overexpressing full-length by osteocytes (25). MEPE is not believed to act on FGF23
DMP1 in bone under control of the 3.6-kilobase (kb) type directly but increases FGF23 levels via inhibition of PHEX
I collagen promoter, although it did completely rescue the enzymatic activity (93). MEPE can be proteolytically
phenotype when overexpressed on a Dmp1-null back- cleaved by cathepsin B or D to release the highly phos-
ground. In contrast, Bhatia et al (88) reported increased phorylated ASARM peptide fragment (94),which in ad-
bone mineral density in transgenic mice that overex- dition to antagonizing PHEX, can also inhibit matrix min-
pressed DMP1 ubiquitously under control of the cytomeg- eralization by binding directly to hydroxyapatite (95, 96).
alovirus promoter. However, in this model, one must take Evidence is growing that modulation of FGF23 expression
into account the fact that the DMP1 is expressed ectopi- by osteocyte proteins is of crucial importance in maintain-
cally in tissues in which it is not normally expressed. ing phosphate homeostasis, and the high levels of expres-
DMP1 loss of function mutations in humans have been sion of all these proteins in osteocytes further confirms
associated with autosomal recessive forms of hypophos- that phosphate regulation is a vital function of these cells.
phatemic rickets (65). These findings provided the first One of the most exciting osteocyte-specific proteins to
evidence that osteocytes may play a major role in the reg- be discovered is sclerostin, the protein product of the
ulation of phosphate homeostasis. Interestingly, osteo- SOST gene (97, 98), which is expressed by mature osteo-
cytes within the osteomalacic bone of Dmp1-null mice cytes (26, 99). Sclerostin is a negative regulator of bone
maintained their expression of alkaline phosphatase, type formation that antagonizes the Wnt/␤-catenin signaling
I collagen, osterix, and E11 and failed to express mature pathway by binding to the Wnt coreceptors, low-density
osteocyte markers, such as sclerostin (65). These studies lipoprotein receptor-related proteins 5 and 6 (LRP5 and
indicated that osteocyte differentiation was arrested at an LRP6) (100). When Wnt ligand binds to LRP5/6, the re-
early stage, therefore suggesting a key role for DMP1 in ceptor is phosphorylated, resulting in the activation of
osteocyte maturation. The effects of DMP1 in the regula- disheveled, which in turn represses glycogen synthase ki-
tion of mineralization and osteocyte maturation appear to nase 3 (GSK3). This results in the release of axin from its
be predominantly due to its role in the regulation of phos- complex with ␤-catenin. The ␤-catenin accumulates and
phate homeostasis because the mineralization defects and translocates to the nucleus, where it binds to transcription
the impairment in osteocyte maturation can be rescued by factors, leading to the activation of Wnt target gene ex-
restoration of normal phosphate homeostasis (38). These pression (reviewed in Ref. 101). However, in the presence
studies also highlight the importance of phosphate regu- of sclerostin, Wnt-receptor interaction is inhibited, and
lation in control of osteocyte differentiation. ␤-catenin is phosphorylated by glycogen synthase kinase
Another protein that is highly expressed in osteocytes is 3 and targeted for ubiquitination and degradation via the
PHEX, which was found to be the target of the osteocyte- proteosome pathway. Studies using loss of function and
specific antibody Mab OB7.3 used to isolate avian osteo- gain of function mouse models of Sost have demonstrated
cytes (89). Mutation of PHEX results in X-linked hy- increased and decreased bone mass, respectively (26, 102).
pophosphatemic rickets (90), and Phex is the mutated Sclerostin expression is decreased by mechanical loading
gene in the Hyp mouse, which is also widely used as a (103), and it was recently reported that the anabolic effects
model of X-linked hypophosphatemic rickets (91). Al- of loading are dependent on this decrease in expression
though the exact function of PHEX remains to be deter- (104). The profound gain of bone mass observed after the
mined, like DMP1, it is known to play a role in phosphate deletion of Sost has led to the development of monoclonal
homeostasis and mineralization of the bone matrix. antibodies directed against sclerostin as a potential ana-
666 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

bolic treatment for osteoporosis. Animal studies and early PTH receptor type 1 (PTH1R) (109), polycystin 1 (PC1)
results from clinical trials look very promising and suggest (110), Mef2C (111), and receptor activator of nuclear fac-
that such antibodies may be an effective therapeutic for tor ␬-B ligand (RANKL) (112, 113).
increasing bone mass (105–107). The Dmp1 promoter has also been used to drive ex-
Microarray comparisons of the gene expression pro- pression of the diphtheria toxin receptor in osteocytes in
files of ex vivo osteoblasts and osteocytes have been per- a murine transgenic model (51). This enabled the condi-
formed to identify other osteocyte-expressed genes that tional ablation of up to 80% of the osteocytes upon in-
may play important roles in bone (21). Increased expres- jection of diphtheria toxin and has provided further in-
sion of genes associated with neurons, such as neuropep- sight into the importance of these cells in processes such as
tide Y, was reported. Further studies have shown that os- mechanotransduction and bone homeostasis, both of
teocyte-expressed neuropeptide Y can inhibit osteoblast which were severely disrupted after osteocyte ablation.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


activity and differentiation (30) and that neuropeptide Y Transgenic mice in which GFP is under control of the 8-kb
expression can be modulated by mechanical loading. Pro- Dmp1 promoter have also been created and have provided
teomic studies have also been used to identify proteins an elegant and valuable new tool for osteocyte research
that are highly expressed by osteocytes. The expression (16). Calvarial cells from these mice were harvested, and
of ORP150, destrin, and macrophage-capping protein the GFP-positive osteocytes were isolated via fluores-
(CapG) were found to be increased in MLO-Y4 cells com- cence-activated cell sorting. The GFP-positive cells were
pared to MC3T3 preosteoblasts, and increased expression then used for gene expression analysis (21).
was confirmed in osteocytes relative to osteoblasts in vivo Before these sophisticated transgenic tools became
(20). Destrin and CapG are believed to play a role in den- available, more traditional methods were used for isola-
drite formation, whereas ORP150 is likely to protect the tion of osteocytes, and such methods still remain impor-
cells from the hypoxic conditions encountered within the tant research tools within the field. These methods include
mineralized bone matrix. the isolation of osteocytes from avian bone by sequential
Figure 3C summarizes some of the main marker genes EDTA and collagenase digestions and their purification
that are differentially expressed during the transition from using the monoclonal antibody OB7.3 (114). Similar di-
an osteoblast to an early and late osteocyte and indicates gestion techniques have also been used on rat calvaria
their temporal expression patterns. The further identifi- (115, 116) and long bones (56) to achieve osteocyte-
cation of such genes is imperative to provide more genetic enriched populations of cells. However, difficulties in ob-
markers for the osteocyte and to provide better tools for taining substantial yields, in addition to maintaining the
defining the steps in the osteocyte differentiation pathway. osteocyte phenotype of the ex vivo cells, have limited the
A greater understanding of the function of these genes is usefulness of these techniques. A recently described
essential to fully elucidate the key roles that the osteocyte method has improved upon these techniques, enabling the
is playing, not only in the maintenance of bone mass, but isolation of greater numbers of osteocytes from mature or
also during endocrine signaling. aged bone by grinding the bone fragments into a fine dust
(117), thereby releasing osteocytes from the mineralized
bone matrix.
V. Tools for Studying Osteocytes Determination of osteocyte function in vitro has been
greatly facilitated by the creation of immortalized osteo-
Although osteocytes remain one of the more challenging cyte-like cell lines. Perhaps the best characterized of these
cells to study due to their inaccessible location within the cell lines are the MLO-Y4 cells, which are representative
mineralized bone matrix, recent advances both in vivo and of an early osteocyte. These cells have been extensively
in vitro have facilitated investigation into their function. studied (118 –120) and their characteristics are outlined in
The development of the Dmp1-Cre mouse line, in which Table 1. However, although MLO-Y4 cells have proved to
Cre recombinase expression is driven by the 10-kb Dmp1 be a very useful tool for studying osteocytes in vitro, they
promoter, has enabled researchers to delete genes specif- cannot provide information on the temporal changes in
ically in Dmp1-expressing cells (50). The authors found gene expression and morphology that occur as an osteo-
that Cre recombinase activity was predominantly con- blast differentiates into an osteocyte. Primary calvarial
fined to osteocytes and odontoblasts with some activity in osteoblasts have been widely used to study terminal os-
a subpopulation of late surface osteoblasts in these mice. teoblast differentiation (121, 122), but the removal of
Although only recently developed, these mice have already these cells from the bone results in a highly heterogeneous
been successfully used to investigate the functions of sev- cell population and may also result in contamination with
eral molecules in osteocytes, including ␤-catenin (108), fibroblasts and osteoclast precursors. Several osteoblastic
doi: 10.1210/er.2012-1026 edrv.endojournals.org 667

Table 1. Characteristics of the Currently Described Osteocyte Cell Lines


Name Origin Characteristics
HOB-01-C1 Human cancellous bone Infected with temperature-sensitive large T antigen, cells proliferate at 34°C but
do not divide at 40°C
Preosteocyte-like phenotype; express low levels of alkaline phosphatase and high
levels of osteocalcin
Synthesize a mineralized matrix (123)
MLO-Y4 C57Bl/6 mouse long bone Express SV40 large T antigen driven by osteocalcin-promoter
Immature osteocyte-like phenotype with dendritic morphology
Low alkaline phosphatase and high osteocalcin expression (120)
Express high levels of the early osteocyte marker E11 (32) but low levels of the
mature osteocyte marker Sost (184, 212)

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


Respond to fluid flow by activating Wnt/␤-catenin signaling (131) and opening
Cx43 hemichannels (163)
Support osteoclast formation and activation in co-culture systems (116) and form
gap junctions with MC3T3-E1 cells (304)
Express primary cilia (137)
MLO-A5 C57Bl/6 mouse long bone Express SV40 large T antigen driven by the osteocalcin promoter
Late osteoblast phenotype and differentiate toward an osteocyte-like phenotype
Synthesize a mineralized matrix, which has a composition similar to normal bone
(124) and is comprised of calcospherulites deposited along collagen fibrils (54)
Express Sost (212), Dmp1, Phex, and Cd44 (36)
IDG-SW3 Long bone cells derived from the Immortomouse IFN-␥ and temperature-sensitive SV40 large T antigen, induces proliferation at
crossed with the 8-kb-Dmp1-GFP mouse (16) 33°C with IFN-␥ but not at 37°C
Synthesize a mineralized matrix
Express GFP under control of the 8-kb Dmp1 promoter
Temporally express early and late osteocyte markers such as E11, Dmp1, Phex,
Mepe, Sost, and Fgf23 (125)
Abbreviation: IFN-␥, interferon-␥.

cell lines have been described that differentiate toward an unloading-induced bone loss (51). These findings further
osteocyte-like phenotype in culture. These include HOB- support the hypothesis that osteocytes are key mechano-
01-C1 preosteocyte cells (123), MLO-A5 late osteoblast transducers in bone.
cells (124), and IDG-SW3 late osteoblast cells (125),
which synthesize and mineralize a type-I collagen extra- A. How osteocytes sense loading
cellular matrix and express known osteocyte markers. When a bone is mechanically loaded, there are several
More information on these cell lines is provided in Table possible stimuli that could be detected by the mechano-
1. These in vitro models form important tools which, sensory osteocyte. These include the physical deformation
alongside in vivo models, are essential for further inves- of the bone matrix itself, the load-induced flow of cana-
tigation into osteocyte differentiation and function. licular fluid through the lacunocanalicular network,
which results in fluid flow shear stress, or electrical stream-
ing potentials that are generated from the flow of the can-
VI. Osteocyte Mechanosensation alicular fluid (which is an ionic solution) past the charged
and Transduction surfaces of the lacunocanalicular walls and/or cell mem-
Two of the earliest functions ascribed to osteocytes were brane. The osteocyte and its dendritic processes are con-
mechanosensation and mechanotransduction. It was first stantly exposed to canalicular fluid that flows through the
recognized by Julius Wolff that bone has the capacity to lacunocanalicular system. It has been proposed that a
adapt to mechanical loading or lack of loading by adding baseline flow of canalicular fluid is driven by the extravas-
or removing bone, thereby modifying bone mass (126). cular pressure, but that on top of this are superimposed
Because of the distribution of osteocytes throughout the rapid alterations in canalicular fluid flow that occur as a
bone matrix and their complex interconnected network, result of intermittent mechanical loading of the bone
early investigators hypothesized that they are the cells re- (127). This results in the cells being exposed to fluid flow
sponsible for sensing mechanical loading or lack of load- shear stress. In vivo, it is difficult to separate out the 3 types
ing. By performing targeted deletion of osteocytes in mice of stimuli because any mechanical loading input will result
expressing the diphtheria toxin receptor specifically in os- in the osteocyte being exposed to all three stimuli, includ-
teocytes, it was shown that these mice were resistant to ing deformation of the bone matrix, fluid flow shear stress
668 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

due to changes in canalicular fluid flow, and associated cells that is associated with the primary cilium. Interest-
streaming potentials. ingly, mice with impaired PC1 function develop osteope-
Almost every cell responds to mechanical loading; how- nia (137), and deletion of PKD1 in osteocytes using the
ever, osteocytes appear to be one of the most sensitive cell 10-kb Dmp1-Cre driver results in reduced bone mass in
types. Osteocytes are more sensitive than osteoblasts and the young skeleton, which subsequently recovers in the
fibroblasts to applied loading in the form of fluid flow adult skeleton. However, these adult mice are less respon-
shear stress and are more sensitive to fluid flow shear stress sive to anabolic load (110). The effects of deleting PC1 are
than to substrate stretching (128, 129). Theoretical mod- reversed by deleting KIF3A, a transport protein known to
els have been used to predict the fluid flow shear stresses play a role in the function of cilia (142).
exerted on osteocyte dendrites as a result of peak physi- Adhesion molecules are proposed to anchor the osteo-
ological loads in vivo and have estimated these to be in the cyte cell body to the lacunar wall and the dendritic pro-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


range of 8 to 30 dynes/cm2 (127). Recently, real-time mea- cesses to the canalicular wall and to potentially interact
surement of load-induced solute transport has been per- with cytoskeletal proteins. These proteins therefore may
formed, and these studies suggested a peak shear stress of play important roles in mechanotransduction. CD44,
5 Pa (approximately 30 dynes/cm2) (130). MLO-Y4 os- which is highly expressed on the osteocyte surface, was the
teocyte-like cells are several orders of magnitude more first adhesion receptor to be described on osteocytes
sensitive to fluid flow shear stress than 2T3 osteoblast-like (29). The actin-bundling protein fimbrin is expressed at
cells (131) and MC3T3 osteoblast cells (132, 133). branches of dendritic processes (58). Both are known to
There has been considerable debate within the field as connect to, and influence, the cell cytoskeleton. Integrins
to which part of the osteocyte is most important in sensing are well known to serve as major receptors/transducers
mechanical strain. Some investigators have proposed that connecting the cytoskeleton to the extracellular matrix in
the osteocyte only senses mechanical load through its den- many cell types (143). It has been proposed that integrins
dritic processes and that the osteocyte cell body is rela- bridge osteocyte processes to their canalicular wall and
tively insensitive to mechanical strain (134, 135). Others that they may play an important role in amplifying the
have proposed that osteocytes sense strain through both strains perceived by osteocyte dendrites as a result of fluid
the cell body and dendritic processes (136), or that the flow (144). Recently, it was found that integrin ␣5␤1 in-
primary cilium, a single hair-like projection found on ev- teracts with Cx43 to mediate the opening of hemichannels
ery cell, is the primary strain-sensing mechanism in the in MLO-Y4 cells in response to mechanical stimulation.
osteocyte (137, 138). There appears to be evidence for all Surprisingly, this interaction appeared to be independent
three mechanisms, and it remains unclear whether the cell of the integrin association with fibronectin and its inter-
body, cell processes, and cilia work separately or in con- action with the extracellular matrix (145). This interac-
junction to sense and transmit mechanical stimuli. Recent tion between integrin ␣5 and Cx43 is important in the
studies using MLO-Y4 cells loaded by fluid flow shear regulation and extracellular release of prostaglandin, an
stress on their dendrites or cell bodies suggest that the essential transducer of the effects of anabolic loading.
glycocalyx, present on the surface of dendritic processes, Therefore, fluid flow shear stress may have 2 major effects
but not the cell body, plays an essential role in mechano- on the osteocyte that are mediated through integrins; the
transduction by dendrites (139). first is the well-known effect of the integrin acting as a
Removing the single primary cilium on either MC3T3 linker between the extracellular matrix and the intracel-
osteoblast or MLO-Y4 osteocyte cells by gene silencing lular cytoskeleton; and the second, novel effect is through
approaches or using treatment with chloral hydrate re- the opening of hemichannels that can release small mol-
duced the amount of prostaglandin released by MLO-Y4 ecules with autocrine and paracrine receptor-mediated
cells in response to fluid flow shear stress and also reduced effects.
increases in cyclooxygenase 2 and the osteoprotegerin It has been shown that the osteocyte lacuna itself acts as
(OPG)/RANKL ratio (138). The primary cilia in bone cells a strain concentrator that amplifies the macroscopic strain
do not appear to mediate calcium flux in response to fluid applied over the whole bone. This strain amplification is
flow; therefore, the mechanism used by primary cilia in strongly affected by the tissue material properties of the
bone cells is distinct from that of kidney cells (138). Re- local perilacunar matrix immediately surrounding the os-
cently, it has been shown that primary cilia signal through teocyte lacuna (136, 146, 147). Using a microstructural
cAMP and adenyl cyclase 6 (140) and can mediate signal- finite element analysis model, these investigators further
ing between osteocytes and mesenchymal stem cells (141). showed that changes in the modulus of the osteocyte cell
It is well known that polycystin 1 (PC1), encoded by the body and dendritic processes had little effect on the max-
gene PKD1, is part of a mechanosensing complex in renal imum strain transmitted to the osteocyte, the average
doi: 10.1210/er.2012-1026 edrv.endojournals.org 669

strain in the cell processes, or the maximum strain in the therefore may play a role in the unique kinetics of calcium
lacuna. In contrast, changing the material properties of the signaling in osteocytes (133).
perilacunar matrix had the greatest impact on the strain Soon after this rapid change in calcium signaling,
transmitted to the osteocyte, with the maximum osteocyte within seconds to minutes, NO, ATP, and prostaglandin
strain relating inversely to the perilacunar tissue modulus. are released. Deleting or inhibiting any one of these three
Therefore, any mechanism that changes the material prop- early small molecules will inhibit bone’s anabolic response
erties of the perilacunar matrix, such as glucocorticoid- to loading (152–156). One of the earliest molecules re-
induced hypomineralization (75), will have significant leased in response to shear stress is NO, which appears to
consequences to mechanosensation by osteocytes. correlate with prostaglandin E2 (PGE2) release from os-
teocytes (128). In bone, NO inhibits resorption and pro-
B. How loading affects osteocyte signaling motes bone formation. Both osteoblasts and osteocytes

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


A true mechanotransducer must be able to convert the release NO in response to mechanical strain or fluid flow
perceived mechanical strain signal into a biological output shear stress (157). NO may also reduce osteocyte apopto-
(ie, a cellular response). Although the precise biochemical sis (158, 159).
pathways have yet to be fully unraveled, considerable ATP is also rapidly released from osteocytes in response
progress has been made toward delineating some of the to extracellular calcium or mechanical stimulation (160,
response pathways in osteocytes that may mediate adap- 161). There is evidence that this occurs through functional
tive responses to mechanical loading and unloading in “hemichannels” that are formed by Cx43 on unopposed
bone. In both primary osteocytes and MLO-Y4 osteocyte- osteocyte cell membranes. The opening of these hemichan-
like cells, fluid flow shear stress increases intracellular cal- nels by mechanical loading appears to provide a mecha-
cium; promotes the release of nitric oxide (NO), ATP, and nism for ATP and NAD⫹ release (162, 163). The P2X7
prostaglandins; induces opening of connexin 43 hemi- nucleotide receptor, an ATP-gated ion channel, may also
channels; and enhances gap junction functions. It has also play a role because its deletion in mice resulted in a 70%
been shown to induce bending of cilia and to initiate sig- reduction in bone anabolic response to applied mechani-
naling pathways such as the Wnt/␤-catenin and protein cal loading, and fluid flow shear stress did not induce
kinase A (PKA) pathways. Fluid flow shear stress also prostaglandin release in cells isolated from these mice
protects osteocytes against apoptosis, activates gene tran- (155). Blockers of P2X7 receptors suppressed prostaglan-
scription and translation, and promotes dendrite elonga- din release, whereas agonists enhanced release in bone
tion. It is still not clear what type of fluid flow shear stress cells, suggesting that the P2X7 receptor is necessary for
occurs physiologically within the lacunocanalicular sys- release of prostaglandin in response to mechanical load.
tem, ie, whether it is steady, pulsatile, oscillatory, or a Prostaglandins may be one of the key effectors of load-
combination of one or more of these. The magnitude, fre- related osteogenic responses. Treatment with prostaglan-
quency, and duration of the fluid flow stimulus in physi- dins in vivo enhances new bone formation in response to
ological conditions are also still unclear. mechanical load (165), and inhibitors of prostaglandin
With regard to the temporal sequence of osteocyte re- production, such as indomethacin, block the effects of
sponses to mechanical loading, one of the earliest events to anabolic loading (152, 166). Fluid flow-induced shear
occur in response to mechanical stimulation is the increase stress stimulates the rapid release of PGE2 by osteocytes
in intracellular calcium. Voltage-operated calcium chan- (163). Interestingly, these authors have shown that PGE2
nels that can be regulated by hormones were shown to be is likely released through Cx43 hemichannels, which serve
expressed in osteoblasts and osteocytes (148, 149). Ex- as a portal for the exit of intracellular PGE2 in osteocytes
pression of voltage-gated potassium (K⫹) channels during stimulated with fluid flow shear stress. This released PGE2
the differentiation from osteoblasts to osteocytes leads to then signals in an autocrine/paracrine manner to the os-
different K⫹ currents during osteoblast to osteocyte tran- teocytes to enhance gap junction function and increase
sition (150). Because of their contribution to maintenance Cx43 expression. It remains to be confirmed whether
of the cell membrane potential and their fast response PGE2 release via Cx43 hemichannels is important for in
within 20 milliseconds, K⫹ channels and other ion chan- vivo load-related responses. So far, in vivo loss of func-
nels may be involved in the earliest initiation of the tion studies suggest that the in vivo role of Cx43 is
mechanical response in osteocytes. The T-type voltage- complex. Cx43 deficiency can either inhibit or enhance
sensitive calcium channel is more highly expressed in os- the bone anabolic response to loading (167, 168), and
teocytes compared to the L-type channel, which is more it has been suggested that the specific promoter used to
highly expressed in osteoblasts (151). The T-type channel delete Cx43 at different stages of the osteoblast lineage
670 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

can affect not only the bone phenotype, but also the chanical loading (32), as are regulators of mineralization
responses to loading (169). and phosphate homeostasis such as PHEX, MEPE, and
As would be surmised, numerous signaling pathways DMP1 (177–179). As mentioned previously, Sost/scleros-
are activated by mechanical loading of osteocytes. One of tin, a marker for the late osteocyte and an inhibitor of
the most important appears to be the Wnt/␤-catenin path- osteoblast function, is down-regulated by anabolic me-
way, important for osteoblast differentiation, prolifera- chanical loading and is increased in response to hind limb
tion, and matrix production. In osteocytes, this pathway unloading (103, 173). Recently, it has been shown that
plays a role in transmitting signals of mechanical loading unloading increases RANKL expression in osteocytes
from osteocytes to cells on the bone surface (reviewed in (113), which may be responsible for the bone loss associ-
Ref. 14). Interestingly, it has been shown that PGE2 re- ated with unloading. It would follow that genes involved
leased by mechanical loading acts through the ␤-catenin in bone formation would be up-regulated in response to

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


pathway to enhance Cx43 expression and gap junction anabolic load and vice versa for unloading. Ideally, it
function (170) and to protect osteocytes from glucocor- would be important to identify genes that are rapidly ac-
ticoid-induced apoptosis (171). tivated in response to anabolic load to deduce the earliest
Deletion of various components of the Wnt/␤-catenin pathways activated by mechanical loading.
pathway has effects on bone responses to loading and un- Overall, there is considerable evidence that osteocytes
loading. In vivo, deletion of LRP5, a major coreceptor for play key roles in mechanosensation and mechanotrans-
Wnt signaling, resulted in mice that showed impaired os- duction in bone, and whereas some of the pathways and
teogenic responses to anabolic loading (172). Negative regulatory mechanisms are beginning to be elucidated,
regulators of the Wnt/␤-catenin pathway such as Dick- there is still much that remains to be determined. A com-
kopf1-related protein 1 (DKK1) and sclerostin are highly plete understanding of these pathways may lead to excit-
expressed in osteocytes (99). Sclerostin is expressed in the ing opportunities for bone anabolic therapeutics because
mature osteocytes, and mechanical loading has been a drug that could mimic the effects of mechanical loading
shown to reduce sclerostin levels in bone (103), whereas or amplify bone anabolic responses to loading would be a
hind limb unloading increases sclerostin expression (173). valuable therapeutic for diseases such as osteoporosis.
It has also been suggested that sclerostin must be down-
regulated in order for the osteogenic response to anabolic
loading to occur (104). Down-regulation of DKK1 and VII. Osteocytes as Orchestrators of Bone
sclerostin may create a permissive environment in which Formation and Resorption
Wnt proteins that are already present can activate the Wnt/
␤-catenin pathway. Whereas targeted deletion of A key feature of osteocytes is their ability to regulate the
␤-catenin using the Dmp1-Cre driver results in dramatic function of both osteoblasts and osteoclasts. It is therefore
bone loss (108), deletion of only 1 allele results in mice becoming clear that in contrast to the historical view of
with a normal skeleton but a completely abrogated re- osteocytes as passive cells, they may actually function as
sponse to anabolic loading (174). Taking all these studies central orchestrators of bone remodeling that can inte-
together, it appears that ␤-catenin plays an important role grate both hormonal and mechanical signals to regulate
not only in osteocyte gap junction function and commu- bone mass. This type of mechanism makes sense because
nication, osteocyte viability, and bone integrity, but also in through the osteocyte’s centralized role in regulating re-
bone responses to loading. sponses to these stimuli, the skeleton would be able to meet
Other signaling pathways are activated in response to not only the demands of its mechanical environment but
mechanical loading and may also cross talk with the Wnt/ also its other demands in relation to calcium and phos-
␤-catenin signaling pathway. Mice lacking the estrogen phate homeostasis.
receptor-␣ (ER-␣) isoform exhibit a defective response to The lacunocanalicular system is the ideal network for
loading (175). Lanyon and co-workers (176) suggest that enabling the transmission of biochemical signals from
the ER-␣ may play a role in shuttling ␤-catenin into the deeply embedded osteocytes to osteoblasts at the bone
nucleus in response to mechanical strain in osteoblasts. surface and thereby allowing osteocytes to influence os-
This may in part explain how estrogen regulates bone mass teoblast activity. Indeed, in vitro studies have shown that
via a functional intersection of the estrogen signaling path- avian osteocytes (180) and MLO-Y4 cells (181) release
way with Wnt/␤-catenin signaling through ER-␣. factors that enhance osteoblast differentiation and alka-
Other genes known to be regulated by loading and by line phosphatase activity. Osteocytes have also been
unloading include E11, a marker for the early osteocyte. shown to induce new bone formation at sites of fracture
This was shown to be up-regulated in response to me- damage by recruiting mesenchymal stem cells via the se-
doi: 10.1210/er.2012-1026 edrv.endojournals.org 671

cretion of osteopontin (182). However, much is still to be studies performed on both MLO-Y4 cells (116) and pri-
learned about the mechanisms by which osteocytes signal mary chick osteocytes (199) show that osteocytes are po-
to osteoblasts. tent supporters of osteoclast formation and activation in
As discussed earlier, signaling molecules such as NO, vitro. In the study by Zhao et al (116), as few as 50
PGE2, and ATP are known to be rapidly released by os- MLO-Y4 cells per well were required to promote robust
teocytes in response to external stimuli such as mechanical osteoclast formation even in the absence of osteotropic
strain, and many of these have direct effects on osteoblasts. factors. Osteoclast precursors express receptor activator
NO generation by endothelial NO synthase (NOS) is be- of nuclear factor ␬-B (RANK) on their cell surface. This
lieved to be the main source of NO from osteocytes (183), receptor binds to its ligand, RANKL, which is expressed
although the inducible form of NOS has been detected in on the surface of cells of the osteoblast lineage, and it is this
osteocytes after mechanical loading (153, 185). Neural binding interaction between RANK and RANKL that pro-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


NOS was also detected in human osteocytes (186). NO motes the differentiation of osteoclast precursors to form
released by osteocytes in vivo has been shown to be im- mature osteoclasts. Importantly, RANKL expression has
portant for bone formation during reloading (153), and in been detected in osteocytes in vivo (200, 201) and
vitro, osteocyte-derived NO inhibits proliferation of early MLO-Y4 cells in vitro (116, 202, 203), consistent with
osteoblasts and promotes osteoblast differentiation (180). their ability to support osteoclast formation. Osteocytes
The effects of PGE2 on osteoblasts and its anabolic effects and MLO-Y4 cells are also known to express another fac-
on bone have also been extensively documented (see Ref. tor required for osteoclast activation, macrophage-colony
187 for review). PGE2 is known to induce bone formation stimulating factor, in addition to OPG, a decoy receptor
(188) and osteoblastic differentiation of primary calvarial for RANKL (116, 204). Deletion of ␤-catenin in osteo-
cell cultures in vitro (189) and to increase overall bone cytes using Dmp1-Cre resulted in decreased expression of
mass in vivo (190, 191). As discussed in Section VI, os- OPG and a subsequent increase in the RANKL/OPG ratio
teogenic responses to mechanical loading are impaired if (108). These mice were characterized by severe osteopo-
production of prostaglandins is inhibited and are en- rosis, which was a consequence of enhanced osteoclast
hanced by treatment with PGE2. activity and excessive bone resorption. The importance of
One of the most important signaling pathways used by osteocytes in regulating osteoclastic activity was recently
osteocytes to regulate bone formation is the Wnt/␤- confirmed in elegant in vivo studies performed by Xiong
catenin pathway. Osteocytes express several modulators et al (113) and Nakashima et al (112) in which RANKL
of Wnt signaling and therefore may regulate osteoblast was deleted specifically in osteocytes. These mouse models
activity via this mechanism. The activation of canonical developed an osteopetrotic phenotype, which led the au-
Wnt signaling in early osteoblasts promotes osteoblast dif- thors to conclude that osteocyte-derived RANKL is essen-
ferentiation and bone formation (192) with opposing ef- tial for normal bone remodeling in adult mice (112, 113).
fects observed when Wnt signaling is disrupted (193). Sev- Therefore, there is a fairly strong body of evidence that
eral osteocyte-secreted Wnt antagonists have been Wnt signaling in osteocytes, via regulation of RANKL/
described, with sclerostin in particular being the focus of OPG, acts as a negative regulator of bone resorption.
many recent studies. In addition to sclerostin, osteocytes Osteocyte death via apoptosis is also thought to result
express the LRP5/6 inhibitor DKK1, which inhibits os- in local bone remodeling via the recruitment of osteoclasts
teoblast differentiation and bone formation (194 –196). to sites of bone damage. Viable osteocytes have been pro-
Similar to sclerostin, DKK1 expression is down-regulated posed to secrete as yet unknown factor(s) that inhibit os-
by mechanical loading in vivo (103), leading to enhanced teoclast activity, and a loss of production of these factors
bone formation. Another Wnt pathway inhibitor ex- due to osteocyte death may release the osteoclasts from
pressed by osteocytes is secreted frizzled-related protein 1 inhibition (205). Consistent with this hypothesis, the tar-
(SFRP1), which is a competitive antagonist of Wnt ligand geted in vivo ablation of osteocytes using the osteocyte-
binding. Loss of SFPR1 expression in vivo results in in- specific diphtheria toxin receptor mouse model was
creased bone mass and mineral density and in vitro en- shown to induce osteoclast formation and activation,
hances osteoblast proliferation and differentiation into os- leading to bone loss (51). Additionally, targeted damage of
teocytes (197). Peak SFRP1 expression is observed in the MLO-Y4 cells cultured in a 3-dimensional collagen gel
immature osteocyte and was decreased in the mature cell, induced osteoclast formation specifically at sites of injury
indicating a role for SFRP1 in the negative regulation of (206) and apoptotic MLO-Y4 cells release HMGB1 (high
cell survival during matrix embedding (198). mobility group box 1), which is chemotactic to osteoclasts
The control of bone remodeling requires regulation of (207). Osteocyte-derived apoptotic bodies have also been
not only osteoblast but also osteoclast activity. Coculture shown to induce osteoclast formation in vitro and in vivo,
672 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

Figure 4. this mouse model, but the net result


was an increase in bone mass. Con-
versely, in a lactating mouse model
in which there is a chronic elevation
in PTHrP, RANKL was shown to be
elevated in osteocytes (215) and
most likely to be responsible for in-
creased osteoclast activity.
Overall, it is becoming clear that it
is not just the osteoblasts and oste-
oclasts that are the critical players in

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


bone remodeling, but that the osteo-
cyte also plays a key role in regulat-
ing the activity of these two cell
types, as summarized in Figure 4.
Moreover, the osteocyte appears to
Figure 4. Osteocyte regulation of bone remodeling. Osteocytes express RANKL and macrophage- be playing a centralized role in inte-
colony stimulating factor (M-CSF) to promote, and OPG and NO to inhibit, osteoclast formation grating mechanical loading and hor-
and activity. Osteocytes also regulate bone formation via the secretion of modulators of the Wnt
signaling pathway. PGE2, NO, and ATP act to activate Wnt signaling, whereas sclerostin, DKK1, monal signals to regulate bone mass
and SFRP1 all inhibit Wnt signaling and subsequent osteoblast activity. Maintenance of this in the skeleton.
balance between resorption and formation by the osteocyte is essential for bone homeostasis.

whereas osteoblast-derived apoptotic bodies had no effect VIII. Osteocyte Life, Death, and in Between
(208). This effect was not mediated by RANKL but was
shown to be TNF-␣-dependent. Not only the healthy, viable osteocyte, but also the dying
As mentioned above, NO signaling can influence bone or dead osteocyte can have important regulatory effects on
mass by increasing bone formation; however, it is also other cell types in bone. Whereas some osteocytes appear
known to inhibit osteoclasts. Conditioned media from me- to sacrifice themselves by apoptosis to initiate bone re-
chanically stimulated chick osteocytes inhibited osteoclast modeling or bone resorption, others remain viable in the
formation and bone resorption in vitro, whereas this effect bone matrix until the death of the organism. Unlike oste-
was blocked upon the addition of an NOS inhibitor (209). oclasts that only live for days and osteoblasts that live for
Osteocyte production of NO therefore appears to pro- weeks, osteocytes are an extremely long-lived cell, surviv-
mote bone formation while at the same time inhibiting ing for up to decades in bone matrix in adults. The life span
bone resorption. of the osteocyte is most likely determined by rates of bone
In addition to their role in mediating adaptive responses turnover, the process by which osteoclasts resorb bone
to mechanical loading, osteocytes can also respond to hor- and osteoblasts replace the resorbed bone. Osteocytes may
monal signals to affect bone formation and resorption. have a half-life of decades if they are located within a bone
The presence of the PTH receptor (PTH1R) has been de- that has a slow turnover rate. The cell is nonmitotic while
tected on the surface of osteocytes as well as osteoblasts it is encased in bone and appears to have developed pro-
(210, 211). Signaling by PTH and PTHrP is known to have tective mechanisms to ensure its survival, especially under
either anabolic or catabolic effects on the skeleton, de- stress conditions such as immobilization, hypoxia, and
pending on whether the administration is intermittent or certain disease conditions. The osteocyte can undergo ne-
continuous. It now appears that osteocytes may be a key crosis, apoptosis, and autophagy, each of which may have
target cell for the anabolic actions of PTH in the skeleton specific regulatory effects on other cells in bone. With age,
because these anabolic effects have been shown to be due, the dying osteocyte appears to undergo a process called
at least partly, to down-regulation of osteocyte Sost ex- micropetrosis, in which the lacuna fills with mineral. Each
pression (212, 213). Constitutive activation of the PTH1R of these states most likely has a significant impact on the
in osteocytes in vivo, under control of the Dmp1 pro- canalicular fluid circulation within the lacunocanalicular
moter, also inhibits Sost expression and dramatically in- network, and on osteocyte cell functions such as mecha-
creases bone formation. This phenotype is dependent on nosensation, signaling, and release of soluble mediators.
increased Wnt signaling (214). Interestingly, increased os- Damaged bone requires replacement. It was proposed
teoclast activity and bone turnover were also observed in that microcracks that occur in bone can damage the os-
doi: 10.1210/er.2012-1026 edrv.endojournals.org 673

teocyte and its processes, thereby inducing the cell to send ceptor modulators, bisphosphonates, calcitonin, CD40 li-
signals of osteoclast recruitment to initiate bone removal gand, and calbindin-D28k (for review, see Ref. 231). In-
and repair. Microdamage and bone fatigue are associated termittent PTH treatment has been shown to increase
with loss of osteocyte integrity (216). Verbogt et al (217) osteocyte density in mice and block osteocyte apoptosis
have also shown that the antiapoptotic factor, B-cell lym- induced by glucocorticoid treatment (232). Increased os-
phoma 2 (BCL-2), is found in osteocytes around the cut- teocyte death is also observed after parathyroidectomy in
ting cone, whereas the proapoptotic factor, BCL-2-asso- patients with secondary hyperparathyroidism (233), sug-
ciated X protein, is found in osteocytes in the path of the gesting a protective role for PTH on osteocytes in vivo.
osteoclast within the cutting cone, suggesting that these Estrogen inhibits etoposide-induced apoptosis (234), and
osteocytes are undergoing programmed cell death. They the estrogen receptor is required for the antiapoptotic ef-
therefore proposed that the dying (apoptotic) osteocytes fect of mechanical stimulation (235). Mechanical loading

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


were sending factors to recruit and direct osteoclasts to in the form of fluid flow shear stress was also shown to
remove the damaged bone. More recently, however, it has protect against glucocorticoid-induced apoptosis through
been shown that the highest expression of osteoclast sup- the release of prostaglandin and subsequent activation of
portive signals is found in a subpopulation of osteocytes the Wnt/␤-catenin pathway (171). A target of the Wnt/␤-
surrounding areas of microdamage. These osteocytes have catenin pathway, OPG also prevents glucocorticoid-
a higher RANKL/OPG ratio than that observed in apo- induced apoptosis (236). Substrate stretching has also
ptotic osteocytes within the areas of microdamage, sug- been shown to protect osteocytes from glucocorticoid-in-
gesting that these viable cells are the ones responsible for duced apoptosis, potentially through activation of FAK,
inducing osteoclastogenesis (218). ERK, and the extracellular matrix-integrin/FAK signaling
It has been shown that osteocytes express markers of that is linked with the Wnt/␤-catenin pathway (237).
apoptosis in response to withdrawal of estrogen (219), to Cx43, the major component of gap junctions and hemi-
oxygen deprivation as occurs during immobilization channels in osteocytes, is also important for osteocyte sur-
(220), and in response to glucocorticoid treatment (221, vival because Cx43 is required for the antiapoptotic effects
222). Increased osteocyte apoptosis was shown to play an of bisphosphonates (238) and deletion of Cx43 in osteo-
important role in the decreased bone strength observed blasts results in animals with increased osteocyte apopto-
with glucocorticoid treatment (223). Further studies have sis (239). Bisphosphonate treatment was shown to reduce
indicated that dexamethasone treatment of MLO-Y4 os- osteocyte apoptosis in response to fatigue loading (240)
teocytes in vitro causes detachment of the cells from the and to protect against glucocorticoid-induced apoptosis
substrate by interfering with focal adhesion kinase (FAK) by transiently increasing ERK phosphorylation (241). A
signaling, resulting in cell death (224). Interestingly, similar effect was also observed with calcitonin and me-
TNF-␣ and IL-1 have both been reported to increase with chanical stimulation (242). Treatment of ovariectomized
estrogen deficiency and also are known inducers of osteo- mice with a pan-caspase inhibitor, QVDOPh, was shown
cyte apoptosis (225). A number of in vitro studies also to inhibit ovariectomy-induced osteocyte apoptosis and
support the concept that the apoptotic osteocyte can sup- reduce osteoclastic bone resorption (243). Inhibitors of
port osteoclast formation. Kogianni et al (208) showed osteocyte apoptosis may therefore prove to be useful ther-
that apoptotic bodies released by MLO-Y4 cells and pri- apeutics for the treatment of diseases of bone loss.
mary osteocytes, but not osteoblasts, supported osteoclast Although much attention was being focused on osteo-
formation. Other groups have shown that serum-starved cyte apoptosis, one group of investigators noted that
MLO-Y4 cells secrete soluble RANKL (226) and that the magnitude of glucocorticoid-induced apoptosis in
damaged MLO-Y4 cell networks embedded in 3-dimen- MLO-Y4 cells was much less than that induced by cyto-
sional gels have increased RANKL and decreased OPG kines such as TNF-␣ and that the same caspase pathways
production (227). Recently, thiazolidinediones have been were not activated (244). They decided to investigate
shown to induce osteocyte apoptosis (228), as does high- whether glucocorticoid treatment could be inducing au-
dose alcohol (229) and methotrexate used for cancer treat- tophagy in these cells (244) and showed that glucocorti-
ment (230). Therefore, a major research focus has been on coid was associated with induction of autophagy-related
osteocyte viability and approaches to prevent osteocyte markers in osteocytes in vitro and in vivo. Autophagy is a
cell death. mechanism in which the cell selectively degrades some of
Searches for inhibitors that would protect osteocytes its own contents in an attempt to maintain viability under
from apoptosis induced by estrogen deprivation or gluco- conditions of stress. It is a tightly regulated process of
corticoid treatment have identified several candidates. lysosomal “self-degradation” in which the cell degrades
These include PTH, estrogen, and selective estrogen re- and recycles nonessential cellular products such as parts of
674 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

the cytoplasm and intracellular organelles to reallocate patients with rickets or osteomalacia (249), uremic osteo-
limited cell resources to processes that are critical for sur- dystrophy (250, 251), and in response to PTH (252, 253).
vival. Autophagy can therefore protect cells from apopto- An increase in the number of lysosomal vesicles was doc-
sis and eventually cell death, thereby preserving viability umented in response to PTH (71, 248, 254). Enlarged
until the stress can be relieved. However, should the stress lacunae have also been described in rats sent into space for
continue or worsen, the outcome can be cell death. Other 22 days (255), hibernating ground squirrels (256), snakes
investigators have also shown glucocorticoid-induced os- (257), and glucocorticoid-treated mice (75). Glycopro-
teocyte autophagy in vivo (245), where it was shown that teins have been detected around the osteocytes of rats
low-dose glucocorticoid induced autophagy and high- treated with parathyroid extract (258) and in lactating rats
dose glucocorticoid induced apoptosis. At the most recent fed with a calcium-free diet (259), and the interpretation
American Society for Bone and Mineral Research meeting, of these data was that the glycoproteins were exposed with

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


it was shown that targeted deletion of the autophagy gene, the removal of mineral.
ATG7, using the Dmp1-Cre driver, which reduced au- Interestingly, Wergedal and Baylink (260) had de-
tophagy in osteocytes, resulted in mice with low bone mass scribed tartrate-resistant acid phosphatase (TRAP) activ-
(246). These data suggest that the long-lived osteocyte ity in osteocytes in 1969, which was criticized as poten-
requires numerous protective mechanisms, one of which is tially being a diffusion artifact from osteoclasts. But this
the capacity to enter the state of autophagy, at least peri- observation was later validated by Nakano et al (261)
odically, to enable it to survive for decades within its en- using in situ hybridization for Trap gene expression. After
closed environment. describing TRAP activity in osteocytes, Baylink went on to
show tetracycline binding to the perilacunar matrix,
which led him to suggest that osteocytes have the ability
IX. Osteocytic Perilacunar Remodeling: An Old not only to remove bone from their perilacunar matrix,
Concept Rediscovered but also to add it back (262). In 1983, Zambonin-Zallone
et al (263) also reported tetracycline labeling in osteocyte
Bone is a porous tissue. Whereas blood vessels and hav- lacunae in egg-laying hens. Although there were numerous
ersian canals are obvious porosities, the osteocyte lacunae reports of osteocytic osteolysis, there had been few reports
are smaller and more numerous, and the canaliculi con- of osteocytes replacing their perilacunar matrix.
tribute to bone porosity to an even greater extent. The Using scanning electron microscopy, gene arrays, and
surface area of the osteocyte lacunocanalicular system is transgenic technology to examine osteocytes in virgin, lac-
estimated to be several orders of magnitude greater than tating, and postlactation mice, it was recently found that
the bone surface area (247). Osteocytes and their dendritic osteocyte lacunar area increases with lactation and returns
processes therefore have access to an extremely large sur- to normal with forced weaning (215). Genes thought to be
face area of bone mineral, and the removal of only a few osteoclast specific, such as Trap and cathepsin K, were
angstroms of mineral from their lacunar and canalicular found to be elevated in osteocytes during lactation and
walls would be expected have significant effects on circu- returned to normal with weaning. PTHrP, which is known
lating, systemic ion levels. Just as osteoclasts and osteo- to be elevated in the circulation during lactation, repro-
blasts remodel the bone surface, osteocytes may have the duced these effects on lacunar enlargement, and this effect
capacity to remodel the surfaces to which they are con- of PTHrP was confirmed to be mediated through the
stantly exposed. PTH1R. This study shows that healthy osteocytes can
Although exciting new functions of osteocytes have both remove and replace their perilacunar matrix during
been discovered within recent years, old functions previ- processes related to normal reproductive function, sug-
ously proposed for osteocytes have also been rediscovered. gesting that osteocytes can potentially play a role in min-
One of these old functions is that of osteocytes being able eral homeostasis during conditions of high calcium de-
to remove their perilacunar matrix, a process that was mand, such as lactation. Interestingly, in this study, no
previously referred to by Belanger et al (248) as “osteo- effect on lacunar size was observed in the hind limbs of tail
cytic osteolysis.” This concept, although proposed in the suspended mice, suggesting that removal of osteocyte peri-
1960s, soon disappeared from the literature, most likely lacunar matrix is not initiated in response to unloading but
because investigators lacked the tools and technology to is targeted by specific signals, such as hormonal
validate the original descriptive observations. Osteocytic regulation.
osteolysis was previously viewed as being a feature of Our work suggests that the osteocyte can not only re-
pathological conditions, especially due to high or contin- move its mineralized matrix but also replace this matrix.
uous PTH. Enlarged lacunae have long been described in We propose that the term “osteocytic osteolysis” be re-
doi: 10.1210/er.2012-1026 edrv.endojournals.org 675

Figure 5. cell cut off from other cells and tissues. How-
ever, the lacunocanalicular system allows for
the flow of canalicular fluid that reflects the
composition of the circulation and exposes the
osteocyte to the hormones and factors that cir-
culate within the blood. A variety of imaging
techniques have demonstrated the connectivity
of osteocyte processes, not only with other
bone cells but also with the vasculature that
permeates the bone matrix (Figure 2, B and C).
Injection of dyes such as procion red into the

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


tail vein of a mouse results in the dye being
distributed via the circulation to the osteocyte
lacunocanalicular system within a matter of
minutes, and studies have shown that mole-
cules up to 70 kDa in size can readily reach the
lacunocanalicular space from the bloodstream
(see Section III). Such connectivity between the
canalicular fluid and vasculature allows osteo-
cytes to be exposed to circulating hormones from
distant tissues, but it would also provide a conduit
for the passage of hormones and other molecular
mediators secreted by osteocytes into the circula-
tion to exert effects on distant target organs. Excit-
ing recent research has discovered the presence of
such osteocyte-produced endocrine factors, with
the result that the osteocytic network should now
be viewed as an endocrine tissue that plays a vital
role in the regulation of phosphate homeostasis.
Figure 5. Endocrine signaling by osteocytes. FGF23 secreted by the osteocyte
regulates serum phosphate (Pi) by down-regulating the expression of
Since its identification in 2000, FGF23 has
sodium/phosphate cotransporters in the kidney. There is also a decrease in 1-␣ emerged as one of the most important osteo-
hydroxylase production by the kidney, resulting in decreased NaPi-IIb in the intestine cyte-secreted endocrine factors. Initially iden-
and reduced Pi uptake. High serum FGF23 has also been linked with an increased
tified in the ventrolateral thalamic nucleus of
risk of left ventricular hypertrophy in the heart, atherosclerosis, and vascular
calcification. However, vascular and soft tissue calcification is increased in Fgf23-null the brain (264), FGF23 was found to be most
mouse models, suggesting that FGF23 can promote and inhibit vascular calcification. highly expressed in bone, predominantly by
FGF23 is also known to negatively regulate the secretion of PTH via the parathyroid. the osteocyte (65, 265–267), and highly ele-
vated in conditions of hypophosphatemia.
served for pathological conditions, whereas the term FGF23 is a 32-kDa member of the FGF family
“perilacunar remodeling” be used for the function of the of proteins that binds to FGF receptors (FGFRs). This
healthy osteocyte such as its function in the healthy lac- binding is dependent on the presence of Klotho. It has been
tating animal. It is interesting to see that an old and ap- shown that Klotho can convert the canonical FGFR,
parently forgotten function for osteocytes has now been FGFR1 (IIIc), into a receptor that is specific for FGF23
resurrected and that the observations of early osteocyte (268) and that a monoclonal antibody targeted against
research pioneers have been validated with modern re- Klotho can prevent activation of FGF23 receptor-medi-
search approaches. ated signaling. FGF23 is known to have effects on the
musculoskeletal system, and increased levels of circulating
FGF23 result in hypophosphatemic disorders; however, it
is the effects of osteocyte-secreted FGF23 beyond the bone
X. The Osteocyte as an Endocrine Cell
that are of particular interest here.
It may initially seem counterintuitive to describe the os- One of the most important targets of FGF23 is the kid-
teocyte as an endocrine cell, considering its location deep ney (Figure 5). Signaling between bone and kidney plays a
within the bone matrix and its appearance as an isolated vital role in the maintenance of serum phosphate levels,
676 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

which is dependent on the circulating levels of FGF23. cells resulted in decreased PTH mRNA expression and re-
FGF23 has been shown to decrease expression of the so- duced the secretion of PTH into the culture media (277).
dium/phosphate cotransporters NaPi-IIa and NaPi-IIc in Similar effects were observed in ex vivo rat parathyroid
the kidney, which are required for renal phosphate reab- glands, where recombinant FGF23 decreased Pth mRNA
sorption (269, 270). This leads to increased urinary ex- expression and secretion of PTH into the media and in vivo,
cretion of phosphate. Additionally, excess FGF23 levels where FGF23 decreased serum PTH levels and Pth mRNA ex-
are known to down-regulate the expression of 1-␣ hy- pression in the parathyroid (278). Increased expression of the
droxylase, which is required for the conversion of 25-hy- FGF23 coreceptor Klotho was also observed in the parathyroid
droxyvitamin D to the active vitamin D metabolite, 1,25- in response to FGF23 administration (278).
dihydroxyvitamin D [1,25(OH)2D] (265). This reduction Recent studies have linked raised levels of circulating
in 1,25(OH)2D levels results in decreased expression of FGF23 to an increased risk of heart disease. Elevated levels of

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


NaPi-IIb in the intestine, therefore reducing phosphate FGF23 were found to be independently associated with left
absorption and leading to hypophosphatemia (271). This ventricular hypertrophy in human population studies (279,
signaling between the osteocyte and the kidney is bidirec- 280). FGF23 also induced hypertrophy in cultured mouse
tional because 1,25(OH)2D induces expression of FGF23 cardiomyocytes, and injection of FGF23 into wild-type mice
by the osteocyte in murine (272) and cell culture models resulted in left ventricular hypertrophy (280). Interestingly,
(125, 272), suggesting a negative feedback system. these effects were found to be Klotho-independent. Increased
In addition to 1,25(OH)2D, several other factors have serum FGF23 has also been linked with impaired vascular
been shown to modulate FGF23 expression and activity. function (279), vascular calcification (281), and increased fat
For example, intake of dietary phosphorous was shown to mass (282), indicating the importance of osteocyte-regulated
regulate serum FGF23 levels in adult mice, with increased proteins in health and disease (as discussed in Section XII).
phosphate supplementation correlating with increases in These findings of altered cardiac and vascular function also
serum phosphate and FGF23 levels (273). Likewise, an identify these tissues as target organs of osteocyte-secreted
increase or decrease in dietary phosphate was shown to FGF23.
result in increased or decreased levels of circulating intact As discussed previously, the osteocyte is known to ex-
FGF23, respectively, in healthy human subjects (274). Re- press several other factors that play a role in the regulation
cent data have also suggested that PTH may directly reg- of phosphate homeostasis. DMP1, PHEX, and MEPE
ulate FGF23 levels. Infusion of PTH for 3 days in adult have well-characterized effects on the skeletal system, as
mice resulted in increased Fgf23 mRNA expression in the demonstrated by loss of function mutations and knockout
calvaria and increased serum FGF23 (275). PTH was also mouse models (65, 91, 283, 284). These secreted proteins
shown to up-regulate expression of Fgf23 mRNA in coordinately regulate FGF23 expression and activity and,
UMR106 cells, although this effect was abolished if by doing so, influence tissues outside of the bone envi-
sclerostin was added to the cultures. In addition, the PKA ronment. DMP1 is a negative regulator of FGF23 ex-
activator forskolin up-regulated expression of FGF23 in pression because osteocytes in the Dmp1-null mouse
these cells, indicating that both the Wnt and PKA pathway express high amounts of FGF23 mRNA and protein,
may modulate the effects of PTH on FGF23. Similar re- compared to wild-type mice, resulting in elevated serum
sults were observed in a recent study by Rhee et al (276), FGF23 levels (65, 266).
in which they generated transgenic mice with constitutive In addition to DMP1, PHEX is also known to inhibit
activation of PTH1R in osteocytes under control of the FGF23. Given that the PHEX protein is an enzyme with
Dmp1 promoter. The authors observed increased FGF23 endopeptidase activity, it was suspected that FGF23 could
expression specifically in osteocytes, and this effect was be proteolytically cleaved by PHEX. However, evidence
abolished by crossing the mice with mice overexpressing has so far suggested that FGF23 is not a direct substrate for
Sost. In vitro studies showed that treatment with PTH, PHEX, but PHEX instead regulates Fgf23 at the transcrip-
PTHrP, and cAMP increased Fgf23 mRNA in osteocyte- tional level (284). The exact mechanism(s) by which
enriched cell cultures, again suggesting the importance of DMP1 and PHEX regulate FGF23 are yet to be deter-
PKA and Wnt signaling in regulating FGF23. mined; however, recent research has suggested that their
As mentioned above, PTH secreted by the parathyroid inhibitory effects are mediated by FGFR signaling (285).
gland increases FGF23 expression in vivo. Interestingly, Dmp1-null and Hyp mouse models show evidence of en-
FGF23 is also able to act reciprocally on the parathyroid hanced FGFR signaling compared to wild-type control
gland to decrease PTH secretion, identifying the parathyroid mice, and the elevated levels of FGF23 present in the bone
gland as another endocrine target of osteocyte signaling. Ad- marrow stromal cells from both mouse models were abol-
dition of recombinant FGF23 to cultured bovine parathyroid ished by inhibition of FGFR signaling.
doi: 10.1210/er.2012-1026 edrv.endojournals.org 677

The effects of MEPE on FGF23 and matrix mineral- pathological FGF23 signaling is cardiac muscle, with in-
ization are dependent on its cleavage to release an acidic creases in FGF23 resulting in left ventricular hypertrophy.
serine aspartate-rich MEPE-associated motif (ASARM) This therefore raises an interesting possibility; can osteo-
from its C terminus. This 19-amino acid ASARM peptide cytes directly influence muscle mass by secreting muscle
is known to be a potent inhibitor of mineralization in vivo regulatory factors? Such interplay between bone and mus-
(94, 286), and Mepe-null mice are characterized by pro- cle seems highly plausible, especially because muscle and
gressive increases in trabecular bone mass and mineral bone mass are tightly correlated throughout life. During
apposition rate with aging (287). Again, the exact mech- organogenesis, muscle and bone develop in close associ-
anisms by which MEPE and the MEPE-ASARM peptide ation from common mesodermal precursors. During ex-
regulate phosphate homeostasis are still to be elucidated; ercise and disuse, changes in muscle and bone mass are
however, the ASARM peptide is known to bind specifi- also closely coupled, and with aging there is a concomitant

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


cally to PHEX in vitro (288) and inhibit PHEX enzymatic loss of both bone and muscle mass. The dogma has been
activity (93), which results in up-regulation of FGF23 ex- that the major effect of muscle on bone is mediated
pression. PHEX can also regulate FGF23 by binding to through mechanical loading of bone via muscle contrac-
MEPE and preventing its proteolytic degradation and the tion during exercise and locomotion. However, it has re-
release of the ASARM peptide (288, 289). In addition, the cently been proposed that muscle, in addition to the skel-
phosphorylated MEPE-ASARM peptide itself is a sub- eton, can act as an endocrine tissue to secrete factors into
strate for PHEX, with cleavage of ASARM by PHEX neu- the circulation that target other tissues. Like osteocytes,
tralizing its activity and restoring mineralization (95). muscle is also a source of circulating signaling factors,
The effects of MEPE can also extend beyond the bone termed “myokines” (296, 297). These myokines include
and may operate independently of FGF23. MEPE is pres- factors such as IGF-I, IL-6, IL-8, IL-15, leukemia inhibi-
ent in the serum of adult humans, suggesting an endocrine
tory factor, FGF21, and follistatin-like 1 (298). Pedersen
function for MEPE, and the levels of MEPE correlate with
(297) has proposed that skeletal muscle, the largest organ
serum phosphate levels (290). Administration of full-
in the body, is an endocrine organ that exerts effects on
length MEPE into a rat model was shown to directly de-
other organs.
crease uptake of phosphate in the proximal tubule and
Previous studies have also demonstrated that skeletal
lead to renal phosphate wasting (291). Likewise, admin-
muscle can secrete factors such as IGF-I, FGF2, and myo-
istration of recombinant MEPE to renal cell cultures in-
statin, which can influence osteoblast activity and differ-
hibited phosphate uptake (291), and short-term infusion
entiation (299, 300). However, whether direct signaling
of MEPE in rats inhibited renal and intestinal phosphate
occurs between muscle and osteocytes was unknown. Re-
absorption independently of serum FGF23, PTH, or
cently, several studies have suggested that such signaling
1,25(OH)2D (292). In addition to the full-length protein,
may indeed play an important physiological role in osteo-
the MEPE-ASARM peptide has also been shown to inhibit
phosphate uptake in vivo (293). MEPE-ASARM is ele- cytes in processes such as regulation of cell viability and
vated in the serum of Hyp mice and was found to accu- response to mechanical stimuli. Jähn et al (301) found that
mulate in the proximal tubules in the Hyp mouse kidney differentiated C2C12 myotubes and primary muscle se-
(294). Overexpression of MEPE in a transgenic mouse crete factors that protect osteocytes against apoptosis due
model was also shown to decrease renal calcification and to glucocorticoids and that muscle contraction enhanced
increase blood vessel formation in the kidney (295), al- factor production. The muscle-secreted factor inhibited
though MEPE expression in this model was also increased glucocorticoid-induced apoptosis through activation of
in the proximal tubules, so the effects cannot be defini- the Wnt/␤-catenin pathway. Lara et al (302) found that
tively attributed to osteocyte-derived MEPE. It is clear that differentiated myotubes and primary muscle produce a
interactions between FGF23, PHEX, MEPE, DMP1, and factor that synergizes with fluid flow shear stress to in-
other SIBLING family members are essential for regulat- crease prostaglandin production by osteocytes. Again, this
ing phosphate levels; however, further studies are required was likely mediated through synergistic activation of the
to fully elucidate how the interplay between these proteins Wnt/␤-catenin pathway (302). Conversely, MLO-Y4 os-
occurs at the genetic and molecular level. teocyte-like cells and primary osteocytes secrete factors
that induce muscle myogenesis in C2C12 pluripotent cells,
and these factors activate the Wnt/␤-catenin pathway
XI. Crosstalk Between Osteocytes
(303). These findings suggest that this pathway plays a
and Muscle Cells
role not only in muscle development but also in muscle
As discussed above, osteocytes can signal to tissues other repair and myogenesis. Two factors produced by osteo-
than bone via secretion of FGF23. One of these targets of cytes, PGE2 and Wnt3a, were found to enhance myogen-
678 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

esis. These early studies provide supporting data that cross bone mass due to increased osteoclast activity (313).
talk occurs between muscle and bone and suggest other RANKL has been shown to be an important mediator of
potential endocrine and/or paracrine functions for osteo- this increase in osteoclast activity, which could be atten-
cytes, but they will require further in vivo validation. uated in a mouse model of secondary hyperparathyroid-
ism by treatment with the endogenous RANKL inhibitor
OPG (314). With recent studies suggesting that osteocytes
XII. Role of Osteocytes in Disease are a major source of RANKL and OPG in vivo (112, 113),
it seems likely that the detrimental effects on bone health
The importance of osteocytes in maintaining bone health observed in hyperparathyroidism are mediated, at least
has been demonstrated by the diseases that occur in their partly, by the osteocyte. In contrast to the effects of con-
absence. A 34% reduction in osteocyte density was ob- tinuous PTH, intermittent PTH treatment is known to be

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


served in cancellous bone of the iliac crest in patients who anabolic and may be partially mediated through down-
presented with fracture when compared to healthy con- regulation of osteocyte-expressed sclerostin (315–317). In
trols, suggesting that osteocytes are important in preserv- humans, it has been reported that patients administered
ing the mechanical properties of bone. These patients did PTH have reduced sclerostin levels (317), and serum
not show an increase in the number of empty lacunae, sclerostin levels in postmenopausal women negatively cor-
however, suggesting that the decrease in osteocyte number relate with PTH (318). Overexpression of Sost in a mouse
was not due to osteocyte apoptosis but was due to a lower model attenuated the anabolic effects of intermittent PTH
rate of osteocyte formation (305). A decrease in endothe- administration (316). However, these animals still
lial NOS and neural NOS expression was also observed in showed a response indicating that the anabolic effect can-
the femoral neck of hip fracture patients, in comparison to not be fully attributed to sclerostin regulation. Sclerostin
nonfracture controls (306, 307). This is likely to result in itself has become a very attractive target for therapeutics
a reduction of NO, a powerful anabolic stimulator (as to increase bone mass. Monoclonal antibodies have been
previously discussed), potentially leading to bone fragility. created to target sclerostin, and their efficacy has been
Apoptosis of osteocytes is commonly observed in pa- tested in both animal and clinical trials. Sclerostin anti-
tients with steroid- and alcohol-induced osteonecrosis of body was shown to protect against bone loss due to hind
the femoral neck (221, 308). Osteocyte death is also ob- limb immobilization in a rat model (107), and it also in-
served after renal transplant, possibly due to the admin- creased bone formation and mineral density in cynomol-
istration of glucocorticoids and other immunosuppressive gus monkey (319) and rat models (320, 321). In a recently
therapeutics (309). There has recently been concern over published human trial, a single treatment with sclerostin
the possibility that therapeutic use of bisphosphonates antibody increased the expression of bone formation
may result in osteonecrosis of the jaw. This has been linked markers such as alkaline phosphatase, decreased markers
to an increase in osteocyte apoptosis in humans (310). In of bone resorption, and led to gains of up to 5.3 and 2.8%
support of this, in a rat experimental model, the admin- in bone mineral density in the vertebrae and hip, respec-
istration of zoledronic acid resulted in osteocyte cell death tively (106). Sclerostin antibody has also shown therapeu-
(311). However, opposing results were observed in mice, tic promise in the treatment of fractures (105) and has
with zoledronic acid having no effect on osteocyte viability alleviated the loss of bone observed after dexamethasone
in the mandible and femur (312). treatment of young mice (322). Off-target effects appear
Abnormalities in osteocyte morphology have been to be minimal, with the only reported issue being a de-
noted in many musculoskeletal diseases. In osteoporotic crease in platelet count and increased megakaryocytes in
bone, disruption of the orientation of the lacunocanalicu- sclerostin antibody-treated rats. These effects were spe-
lar system is observed, with decreased connectivity be- cies-specific, with no reactivity between the antibodies and
tween neighboring osteocytes. Osteoarthritic bone is char- human or monkey platelets or megakaryocytes (323).
acterized by a decrease in osteocyte connectivity; however, As discussed in Section X, the effects of osteocyte sig-
the orientation of the processes is unchanged. In contrast, naling are not merely confined to the bone microenviron-
osteocytes in osteomalacic bone display high connectivity ment but can involve other tissues such as the kidney and
but with disorientation of the dendrites (59). Such disrup- the cardiovascular system. FGF23 expression is known to
tion of the lacunocanalicular system will affect osteocyte be regulated by DMP1, PHEX, and MEPE, which are all
signaling, likely leading to alterations in the bone mechan- predominantly expressed by osteocytes. Elevated levels of
ical properties. circulating FGF23 lead to increased renal phosphate ex-
Hyperparathyroidism has a profound effect on the skel- cretion and subsequent hypophosphatemia and osteoma-
etal system. Excessive PTH secretion results in a loss of lacia (270, 324). In chronic kidney disease, serum levels of
doi: 10.1210/er.2012-1026 edrv.endojournals.org 679

FGF23 are increased, particularly in the later stages of the may have on other tissues. It also raises the question of
disease (325, 326). Treatment with FGF23 antibody re- which other cells and tissues could be communicating with
stored serum phosphate levels in the Hyp mouse model the osteocyte. The extensive connectivity of the lacuno-
and corrected the osteomalacic phenotype (327). Like- canalicular system with the vasculature permits the trans-
wise, treatment of Dmp1-null mice with FGF23 antibody mission of signals to and from osteocytes from tissues
ameliorated the bone defects observed in these mice by throughout the body, and it is highly likely that other
increasing levels of circulating phosphate (38). In addition targets of osteocyte signaling will be discovered in the near
to effects on the musculoskeletal system, high serum future.
FGF23 levels have been linked with atherosclerosis (328), The osteocyte is gaining increasing interest as a target
ventricular hypertrophy (329, 330), increased risk of car- of therapeutics to increase bone mass. Clinical and animal
diovascular disease (331), and vascular calcification trials have shown that using monoclonal antibodies di-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


(281). Treatments targeting FGF23 would therefore ap- rected against sclerostin has a potent bone anabolic effect.
pear to offer significant therapeutic potential and not Future research may yield additional osteocyte secreted
merely for bone homeostasis. factors, which may have therapeutic potential in disorders
Decreased bone mass is a common feature in inflam- such as sarcopenia and chronic kidney failure, given the
matory bowel disorders, particularly in children (332, cross talk between these organs. Clearly, the osteocyte
333), and it was recently reported that the bone loss ob- must now be considered not only in the context of its
served in Crohn’s disease was associated with increased immediate microenvironment but also in its far-reaching
osteocyte death and decreased bone remodeling (334). endocrine effects beyond the bone—therefore, adding to a
The role of osteocytes in cancer-induced bone disease growing list of functions of these fascinating, dynamic
has, so far, been relatively unexplored. However, it was cells.
discovered recently that there is an increase in osteocyte
apoptosis, and subsequently a decrease in viable osteo-
cytes, in the bones of patients suffering from multiple my- Acknowledgments
eloma (335). This resulted in an increase in osteoclast for- Address all correspondence and requests for reprints to: Lynda F. Bone-
mation and osteolytic lesions. A recent study has also wald, PhD, Department of Oral and Craniofacial Sciences, School of
suggested that circulating levels of sclerostin are elevated Dentistry, University of Missouri-Kansas City, 650 East 25th Street,
in patients suffering from multiple myeloma and correlate Kansas City, Missouri 64108. E-mail: Bonewaldl@umkc.edu.
This work was supported by grants from the National Institutes of
with decreased bone mass (336). Other Wnt signaling in-
Health (PO1 AR46798 and PO1 AG039355 to L.F.B. and R21
hibitors secreted by osteocytes, such as DKK1 and SFRP1, AR054449, RO1 ARO51517, and S10 RR027668 to S.L.D.).
are also known to be involved in myeloma-induced bone Disclosure Summary: The authors have nothing to disclose.
destruction (86, 164). The importance of the osteocyte as
a source of these factors in multiple myeloma, however,
remains to be determined. References

1. Beno T, Yoon YJ, Cowin SC, Fritton SP. Estimation of


bone permeability using accurate microstructural measure-
XIII. Summary and Perspective ments. J Biomech. 2006;39(13):2378 –2387.
2. Franz-Odendaal TA, Hall BK, Witten PE. Buried alive:
It is becoming clear that the osteocyte is a far more active how osteoblasts become osteocytes. Dev Dyn. 2006;
and functional cell than dogma has previously suggested. 235(1):176 –190.
Recent research has further proved many of the suspected 3. Bonewald LF. The amazing osteocyte. J Bone Miner Res.
functions of osteocytes, such as mechanosensation and 2011;26(2):229 –238.
4. Dallas SL, Bonewald LF. Dynamics of the transition from
regulation of bone remodeling, in addition to suggesting
osteoblast to osteocyte. Ann NY Acad Sci. 2010;1192(1):
novel functions, such as the ability to act as an endocrine 437– 443.
organ. The potential of the osteocyte to influence cells and 5. Manolagas SC. Birth and death of bone cells: basic regu-
tissues far beyond the bone microenvironment suggests latory mechanisms and implications for the pathogenesis
that we may only be “scratching the surface” of what the and treatment of osteoporosis. Endocr Rev. 2000;21(2):
115–137.
osteocyte is capable of. Signaling between osteocytes and
6. Jilka RL, Weinstein RS, Bellido T, Parfitt AM, Manolagas
the parathyroid, kidney, cardiac and skeletal muscle in- SC. Osteoblast programmed cell death (apoptosis): mod-
dicates the importance of the endocrine function of osteo- ulation by growth factors and cytokines. J Bone Miner Res.
cytes and the impact that maintaining osteocyte viability 1998;13(5):793– 802.
680 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

7. Holtrop ME. Light and electron microscopical structure of bone remodeling. J Electron Microsc (Tokyo). 2009;58(6):
bone forming cells. In: Hall BK, ed. Bone. Vol. 1: The Os- 381–392.
teoblast and Osteocyte. Caldwell, NJ: The Telford Press; 25. Igarashi M, Kamiya N, Ito K, Takagi M. In situ localization
1990:1– 40. and in vitro expression of osteoblast/osteocyte factor 45
8. Nefussi JR, Sautier JM, Nicolas V, Forest N. How osteo- mRNA during bone cell differentiation. Histochem J.
blasts become osteocytes: a decreasing matrix forming pro- 2002;34(5):255–263.
cess. J Biol Buccale. 1991;19(1):75– 82. 26. Winkler DG, Sutherland MK, Geoghegan JC, et al. Osteo-
9. Nijweide PJ, van der Plas A, Scherft JP. Biochemical and cyte control of bone formation via sclerostin, a novel BMP
histological studies on various bone cell preparations. Cal- antagonist. EMBO J. 2003;22(23):6267– 6276.
cif Tissue Int. 1981;33(5):529 –540. 27. Guo D, Keightley JA, Barragan L, Zhao J, Guthrie J, Bone-
10. Palumbo C. A three-dimensional ultrastructural study of wald LF. Identification of proteins involved in cytoskeletal
osteoid-osteocytes in the tibia of chick embryos. Cell Tissue rearrangement, anti-hypoxia and membrane channels in
Res. 1986;246(1):125–131. osteocytes over osteoblasts. J Bone Miner Res. 2006;

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


11. Palumbo C, Palazzini S, Zaffe D, Marotti G. Osteocyte 21(suppl 1):S168.
differentiation in the tibia of newborn rabbit: an ultra- 28. Yang W, Harris MA, Heinrich JG, Guo D, Bonewald LF,
structural study of the formation of cytoplasmic processes. Harris SE. Gene expression signatures of a fibroblastoid
Acta Anat (Basel). 1990;137(4):350 –358. preosteoblast and cuboidal osteoblast cell model compared
12. Zhao W, Byrne MH, Wang Y, Krane SM. Osteocyte and to the MLO-Y4 osteocyte cell model. Bone. 2009;44(1):
osteoblast apoptosis and excessive bone deposition accom- 32– 45.
pany failure of collagenase cleavage of collagen. J Clin In- 29. Hughes DE, Salter DM, Simpson R. CD44 expression in
vest. 2000;106(8):941–949. human bone: a novel marker of osteocytic differentiation.
13. Holmbeck K, Bianco P, Pidoux I, et al. The metalloprotei- J Bone Miner Res. 1994;9(1):39 – 44.
nase MT1-MMP is required for normal development and 30. Igwe JC, Jiang X, Paic F, et al. Neuropeptide Y is expressed
maintenance of osteocyte processes in bone. J Cell Sci. by osteocytes and can inhibit osteoblastic activity. J Cell
2005;118:147–156. Biochem. 2009;108(3):621– 630.
14. Bonewald LF, Johnson ML. Osteocytes, mechanosensing 31. Wetterwald A, Hoffstetter W, Cecchini MG, et al. Char-
and Wnt signaling. Bone. 2008;42(4):606 – 615. acterization and cloning of the E11 antigen, a marker ex-
15. Bonewald LF. Osteocytes as dynamic multifunctional cells. pressed by rat osteoblasts and osteocytes. Bone. 1996;
Ann NY Acad Sci. 2007;1116:281–290. 18(2):125–132.
16. Kalajzic I, Braut A, Guo D, et al. Dentin matrix protein 1 32. Zhang K, Barragan-Adjemian C, Ye L, et al. E11/gp38
expression during osteoblastic differentiation, generation selective expression in osteocytes: regulation by mechani-
of an osteocyte GFP-transgene. Bone. 2004;35(1):74 – 82. cal strain and role in dendrite elongation. Mol Cell Biol.
17. Kalajzic I, Kalajzic Z, Kaliterna M, et al. Use of type I 2006;26(12):4539 – 4552.
collagen green fluorescent protein transgenes to identify 33. Lu Y, Yuan B, Qin C, et al. The biological function of
subpopulations of cells at different stages of the osteoblast DMP-1 in osteocyte maturation is mediated by its 57-kDa
lineage. J Bone Miner Res. 2002;17(1):15–25. C-terminal fragment. J Bone Miner Res. 2011;26(2):331–
18. Kalajzic Z, Liu P, Kalajzic I, et al. Directing the expression 340.
of a green fluorescent protein transgene in differentiated 34. Atkins GJ, Rowe PS, Lim HP, et al. Sclerostin is a locally
osteoblasts: comparison between rat type I collagen and rat acting regulator of late-osteoblast/preosteocyte differenti-
osteocalcin promoters. Bone. 2002;31(6):654 – 660. ation and regulates mineralization through a MEPE-
19. Billiard J, Moran RA, Whitley MZ, et al. Transcriptional ASARM-dependent mechanism. J Bone Miner Res. 2011;
profiling of human osteoblast differentiation. J Cell 26(7):1425–1436.
Biochem. 2003;89(2):389 – 400. 35. Hirao M, Hashimoto J, Yamasaki N, et al. Oxygen tension
20. Guo D, Keightley A, Guthrie J, Veno PA, Harris SE, Bone- is an important mediator of the transformation of osteo-
wald LF. Identification of osteocyte-selective proteins. Pro- blasts to osteocytes. J Bone Miner Metab. 2007;25(5):
teomics. 2010;10(20):3688 –3698. 266 –276.
21. Paic F, Igwe JC, Nori R, et al. Identification of differentially 36. Prideaux M, Loveridge N, Pitsillides AA, Farquharson C.
expressed genes between osteoblasts and osteocytes. Bone. Extracellular matrix mineralization promotes E11/gp38
2009;45:682– 692. glycoprotein expression and drives osteocytic differentia-
22. Schulze E, Witt M, Kasper M, Lowik CW, Funk RH. Im- tion. PLoS One. 2012;7(5):e36786.
munohistochemical investigations on the differentiation 37. Irie K, Ejiri S, Sakakura Y, Shibui T, Yajima T. Matrix
marker protein E11 in rat calvaria, calvaria cell culture and mineralization as a trigger for osteocyte maturation. J His-
the osteoblastic cell line ROS 17/2.8. Histochem Cell Biol. tochem Cytochem. 2008;56(6):561–567.
1999;111(1):61– 69. 38. Zhang R, Lu Y, Ye L, et al. Unique roles of phosphorus in
23. Toyosawa S, Shintani S, Fujiwara T, et al. Dentin matrix endochondral bone formation and osteocyte maturation.
protein 1 is predominantly expressed in chicken and rat J Bone Miner Res. 2011;26(5):1047–1056.
osteocytes but not in osteoblasts. J Bone Miner Res. 2001; 39. Marotti G. The structure of bone tissues and the cellular
16(11):2017–2026. control of their deposition. Ital J Anat Embryol. 1996;
24. Ubaidus S, Li M, Sultana S, et al. FGF23 is mainly synthe- 101(4):25–79.
sized by osteocytes in the regularly distributed osteocytic 40. Dallas SL, Veno PA, Bonewald LF, Rowe DW, Kalajzic I.
lacunar canalicular system established after physiological Dynamic imaging of fluorescently tagged osteoblast and
doi: 10.1210/er.2012-1026 edrv.endojournals.org 681

osteocyte populations integrates mineralization dynamics tribution of actin-binding proteins. J Bone Miner Res.
with osteoblast to osteocyte transition. J Bone Miner Res. 2004;19(3):471– 478.
2007;22(suppl 1):S13. 58. Tanaka-Kamioka K, Kamioka H, Ris H, Lim SS. Osteocyte
41. Veno PA, Lu Y, Kamel S, Feng J, Bonewald LF, Dallas SL. shape is dependent on actin filaments and osteocyte pro-
A membrane targeted GFP selectively expressed in osteo- cesses are unique actin-rich projections. J Bone Miner Res.
cytes reveals cell and membrane dynamics in living osteo- 1998;13(10):1555–1568.
cytes. J Bone Miner Res. 2010;25(suppl 1):S56. 59. Knothe Tate ML, Adamson JR, Tami AE, Bauer TW. The
42. Candeliere GA, Liu F, Aubin JE. Individual osteoblasts in osteocyte. Int J Biochem Cell Biol. 2004;36(1):1– 8.
the developing calvaria express different gene repertoires. 60. Bonewald LF. Mechanosensation and transduction in os-
Bone. 2001;28(4):351–361. teocytes. Bonekey Osteovision. 2006;3(10):7–15.
43. Liu F, Malaval L, Aubin JE. The mature osteoblast phe- 61. Doty SB. Morphological evidence of gap junctions be-
notype is characterized by extensive plasticity. Exp Cell tween bone cells. Calcif Tissue Int. 1981;33(5):509 –512.
Res. 1997;232(1):97–105. 62. Bennett MV, Goodenough DA. Gap junctions, electro-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


44. Imai S, Heino TJ, Hienola A, et al. Osteocyte-derived HB- tonic coupling, and intercellular communication. Neurosci
GAM (pleiotrophin) is associated with bone formation and Res Program Bull. 1978;16(3):1– 486.
mechanical loading. Bone. 2009;44(5):785–794. 63. Kamioka H, Honjo T, Takano-Yamamoto T. A three-di-
45. Suzuki R, Domon T, Wakita M. Some osteocytes released mensional distribution of osteocyte processes revealed by
from their lacunae are embedded again in the bone and not the combination of confocal laser scanning microscopy
engulfed by osteoclasts during bone remodeling. Anat Em- and differential interference contrast microscopy. Bone.
bryol (Berl). 2000;202(2):119 –128. 2001;28(2):145–149.
46. van der Plas A, Aarden EM, Feijen JH, et al. Characteristics 64. Veno PA, Nicolella DP, Kalajzic I, Rowe DW, Bonewald
and properties of osteocytes in culture. J Bone Miner Res. LF, Dallas SL. Dynamic imaging in living calvaria reveals
1994;9(11):1697–1704. the motile properties of osteoblasts and osteocytes and sug-
47. Suzuki R, Domon T, Wakita M, Akisaka T. The reaction gests heterogeneity of osteoblasts in bone. J Bone Miner
of osteoclasts when releasing osteocytes from osteocytic
Res. 2007;22(suppl 1):S13.
lacunae in the bone during bone modeling. Tissue Cell.
65. Feng JQ, Ward LM, Liu S, et al. Loss of DMP1 causes
2003;35(3):189 –197.
rickets and osteomalacia and identifies a role for osteocytes
48. Bronckers AL, Goei W, Luo G, et al. DNA fragmentation
in mineral metabolism. Nat Genet. 2006;38(11):1310 –
during bone formation in neonatal rodents assessed by
1315.
transferase-mediated end labeling. J Bone Miner Res.
66. Knothe Tate ML, Niederer P, Knothe U. In vivo tracer
1996;11(9):1281–1291.
transport through the lacunocanalicular system of rat bone
49. Elmardi AS, Katchburian MV, Katchburian E. Electron
in an environment devoid of mechanical loading. Bone.
microscopy of developing calvaria reveals images that sug-
1998;22(2):107–117.
gest that osteoclasts engulf and destroy osteocytes during
67. Wang L, Ciani C, Doty SB, Fritton SP. Delineating bone’s
bone resorption. Calcif Tissue Int. 1990;46(4):239 –245.
interstitial fluid pathway in vivo. Bone. 2004;34(3):499 –
50. Lu Y, Xie Y, Zhang S, Dusevich V, Bonewald LF, Feng JQ.
DMP1-targeted Cre expression in odontoblasts and osteo- 509.
cytes. J Dent Res. 2007;86(4):320 –325. 68. Fritton SP, Weinbaum S. Fluid and solute transport in
51. Tatsumi S, Ishii K, Amizuka N, et al. Targeted ablation of bone: flow-induced mechanotransduction. Annu Rev
osteocytes induces osteoporosis with defective mechano- Fluid Mech. 2009;41:347–374.
transduction. Cell Metab. 2007;5(6):464 – 475. 69. Weinbaum S, Duan Y, Thi MM, You L. An integrative
52. Palumbo C, Ferretti M, Marotti G. Osteocyte dendrogen- review of mechanotransduction in endothelial, epithelial
esis in static and dynamic bone formation: an ultrastruc- (renal) and dendritic cells (osteocytes). Cell Mol Bioeng.
tural study. Anat Rec A Discov Mol Cell Evol Biol. 2004; 2011;4(4):510 –537.
278(1):474 – 480. 70. Knothe Tate ML. “Whither flows the fluid in bone?” An
53. Palumbo C, Palazzini S, Marotti G. Morphological study osteocyte’s perspective. J Biomech. 2003;36(10):1409 –
of intercellular junctions during osteocyte differentiation. 1424.
Bone. 1990;11(6):401– 406. 71. Belanger LF. Osteocytic osteolysis. Calcif Tissue Res.
54. Barragan-Adjemian C, Nicolella D, Dusevich V, Dallas 1969;4(1):1–12.
MR, Eick JD, Bonewald LF. Mechanism by which 72. Belanger LF. Osteocytic osteolysis in a cretaceous reptile.
MLO-A5 late osteoblasts/early osteocytes mineralize in Rev Can Biol. 1977;36(1):71–73.
culture: similarities with mineralization of lamellar bone. 73. Doty SB, Nunez EA. Activation of osteoclasts and the re-
Calcif Tissue Int. 2006;79(5):340 –353. population of bone surfaces following hibernation in the
55. Dudley HR, Spiro D. The fine structure of bone cells. J Bio- bat, Myotis lucifugus. Anat Rec. 1985;213(4):481– 495.
phys Biochem Cytol. 1961;11:627– 649. 74. Steinberg B, Singh IJ, Mitchell OG. The effects of cold-
56. Gu G, Nars M, Hentunen TA, Metsikko K, Vaananen HK. stress. Hibernation, and prolonged inactivity on bone dy-
Isolated primary osteocytes express functional gap junc- namics in the golden hamster, Mesocricetus auratus. J Mor-
tions in vitro. Cell Tissue Res. 2006;323(2):263–271. phol. 1981;167(1):43–51.
57. Kamioka H, Sugawara Y, Honjo T, Yamashiro T, Takano- 75. Lane NE, Yao W, Balooch M, et al. Glucocorticoid-treated
Yamamoto T. Terminal differentiation of osteoblasts to mice have localized changes in trabecular bone material
osteocytes is accompanied by dramatic changes in the dis- properties and osteocyte lacunar size that are not observed
682 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

in placebo-treated or estrogen-deficient mice. J Bone Miner Gy and Hyp mice provide mouse models for X-linked hy-
Res. 2006;21(3):466 – 476. pophosphatemia. Hum Mol Genet. 1997;6(2):165–171.
76. Okada S, Yoshida S, Ashrafi SH, Schraufnagel DE. The 92. Liu S, Tang W, Zhou J, Vierthaler L, Quarles LD. Distinct
canalicular structure of compact bone in the rat at different roles for intrinsic osteocyte abnormalities and systemic fac-
ages. Microsc Microanal. 2002;8(2):104 –115. tors in regulation of FGF23 and bone mineralization in
77. Hantusch B, Kalt R, Krieger S, Puri C, Kerjaschki D. Sp1/ Hyp mice. Am J Physiol Endocrinol Metab. 2007;293(6):
Sp3 and DNA-methylation contribute to basal transcrip- E1636 –E1644.
tional activation of human podoplanin in MG63 versus 93. Liu S, Rowe PS, Vierthaler L, Zhou J, Quarles LD. Phos-
Saos-2 osteoblastic cells. BMC Mol Biol. 2007;8:20. phorylated acidic serine-aspartate-rich MEPE-associated
78. Kunita A, Kashima TG, Ohazama A, Grigoriadis AE, Fu- motif peptide from matrix extracellular phosphoglycopro-
kayama M. Podoplanin is regulated by AP-1 and promotes tein inhibits phosphate regulating gene with homologies to
platelet aggregation and cell migration in osteosarcoma. endopeptidases on the X-chromosome enzyme activity. J
Am J Pathol. 2011;179(2):1041–1049. Endocrinol. 2007;192(1):261–267.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


79. Martin-Villar E, Yurrita MM, Fernandez-Munoz B, Quin- 94. Rowe PS, Kumagai Y, Gutierrez G, et al. MEPE has the
tanilla M, Renart J. Regulation of podoplanin/PA2.26 an- properties of an osteoblastic phosphatonin and minhibin.
tigen expression in tumour cells. Involvement of calpain- Bone. 2004;34(2):303–319.
mediated proteolysis. Int J Biochem Cell Biol. 2009;41(6): 95. Addison WN, Nakano Y, Loisel T, Crine P, McKee MD.
1421–1429. MEPE-ASARM peptides control extracellular matrix min-
80. Ohizumi I, Harada N, Taniguchi K, et al. Association of eralization by binding to hydroxyapatite: an inhibition reg-
CD44 with OTS-8 in tumor vascular endothelial cells. ulated by PHEX cleavage of ASARM. J Bone Miner Res.
Biochim Biophys Acta. 2000;1497(2):197–203. 2008;23(10):1638 –1649.
81. Sprague LW, Heinzman U, Atkinson MJ. Phenotypic 96. Boskey AL, Chiang P, Fermanis A, et al. MEPE’s diverse
changes following over-expression of sense or antisense effects on mineralization. Calcif Tissue Int. 2010;86(1):
E11 cDNA in ROS 17/2.8 cells. J Bone Miner Res. 1996; 42– 46.
97. Balemans W, Ebeling M, Patel N, et al. Increased bone
11(suppl 1):S132.
density in sclerosteosis is due to the deficiency of a novel
82. Scholl FG, Gamallo C, Vilaró S, Quintanilla M. Identifi-
secreted protein (SOST). Hum Mol Genet. 2001;10(5):
cation of PA2.26 antigen as a novel cell-surface mucin-type
537–543.
glycoprotein that induces plasma membrane extensions
98. Brunkow ME, Gardner JC, Van Ness J, et al. Bone dys-
and increased motility in keratinocytes. J Cell Sci. 1999;
plasia sclerosteosis results from loss of the SOST gene prod-
112:4601– 4613.
uct, a novel cystine knot-containing protein. Am J Hum
83. Martin-Villar E, Megias D, Castel S, Yurrita MM, Vilaro
Genet. 2001;68(3):577–589.
S, Quintanilla M. Podoplanin binds ERM proteins to ac-
99. Poole KE, van Bezooijen RL, Loveridge N, et al. Sclerostin
tivate RhoA and promote epithelial-mesenchymal transi-
is a delayed secreted product of osteocytes that inhibits
tion. J Cell Sci. 2006;119:4541– 4553.
bone formation. FASEB J. 2005;19(13):1842–1844.
84. Schacht V, Ramirez MI, Hong YK, et al. T1␣/podoplanin
100. Li X, Zhang Y, Kang H, et al. Sclerostin binds to LRP5/6
deficiency disrupts normal lymphatic vasculature forma- and antagonizes canonical Wnt signaling. J Biol Chem.
tion and causes lymphedema. EMBO J. 2003;22(14): 2005;280(20):19883–19887.
3546 –3556. 101. Ott SM. Sclerostin and Wnt signaling—the pathway to
85. Feng JQ, Zhang J, Dallas SL, et al. Dentin matrix protein bone strength. J Clin Endocrinol Metab. 2005;90(12):
1, a target molecule for Cbfa1 in bone, is a unique bone 6741– 6743.
marker gene. J Bone Miner Res. 2002;17(10):1822–1831. 102. Li X, Ominsky MS, Niu QT, et al. Targeted deletion of the
86. Heath DJ, Chantry AD, Buckle CH, et al. Inhibiting Dick- sclerostin gene in mice results in increased bone formation
kopf-1 (Dkk1) removes suppression of bone formation and and bone strength. J Bone Miner Res. 2008;23(6):860 –
prevents the development of osteolytic bone disease in mul- 869.
tiple myeloma. J Bone Miner Res. 2009;24(3):425– 436. 103. Robling AG, Niziolek PJ, Baldridge LA, et al. Mechanical
87. Feng JQ, Huang H, Lu Y, et al. The Dentin matrix protein stimulation of bone in vivo reduces osteocyte expression of
1 (Dmp1) is specifically expressed in mineralized, but not Sost/sclerostin. J Biol Chem. 2008;283(9):5866 –5875.
soft, tissues during development. J Dent Res. 2003;82(10): 104. Tu X, Rhee Y, Condon KW, et al. Sost downregulation and
776 –780. local Wnt signaling are required for the osteogenic re-
88. Bhatia A, Albazzaz M, Espinoza Orias AA, et al. Overex- sponse to mechanical loading. Bone. 2012;50(1):209 –
pression of DMP1 accelerates mineralization and alters 217.
cortical bone biomechanical properties in vivo. J Mech Be- 105. Ominsky MS, Li C, Li X, et al. Inhibition of sclerostin by
hav Biomed Mater. 2012;5(1):1– 8. monoclonal antibody enhances bone healing and improves
89. Westbroek I, De Rooij KE, Nijweide PJ. Osteocyte-specific bone density and strength of nonfractured bones. J Bone
monoclonal antibody MAb OB7.3 is directed against Phex Miner Res. 2011;26(5):1012–1021.
protein. J Bone Miner Res. 2002;17(5):845– 853. 106. Padhi D, Jang G, Stouch B, Fang L, Posvar E. Single-dose,
90. A gene (PEX) with homologies to endopeptidases is mu- placebo-controlled, randomized study of AMG 785, a
tated in patients with X-linked hypophosphatemic rickets. sclerostin monoclonal antibody. J Bone Miner Res. 2011;
The HYP Consortium. Nat Genet. 1995;11(2):130 –136. 26(1):19 –26.
91. Strom TM, Francis F, Lorenz B, et al. Pex gene deletions in 107. Tian X, Jee WS, Li X, Paszty C, Ke HZ. Sclerostin antibody
doi: 10.1210/er.2012-1026 edrv.endojournals.org 683

increases bone mass by stimulating bone formation and 126. Wolff J. Das Gesetz der Transformation der Knochen. Ber-
inhibiting bone resorption in a hindlimb-immobilization lin: A Hirschwald; 1892.
rat model. Bone. 2011;48(2):197–201. 127. Weinbaum S, Cowin SC, Zeng Y. A model for the excita-
108. Kramer I, Halleux C, Keller H, et al. Osteocyte Wnt/␤- tion of osteocytes by mechanical loading-induced bone
catenin signaling is required for normal bone homeostasis. fluid shear stresses. J Biomech. 1994;27(3):339 –360.
Mol Cell Biol. 2010;30(12):3071–3085. 128. Klein-Nulend J, van der Plas A, Semeins CM, et al. Sensi-
109. Powell WF Jr, Barry KJ, Tulum I, et al. Targeted ablation tivity of osteocytes to biomechanical stress in vitro. FASEB
of the PTH/PTHrP receptor in osteocytes impairs bone J. 1995;9(5):441– 445.
structure and homeostatic calcemic responses. J Endocri- 129. Klein-Nulend J, Semeins CM, Ajubi NE, Nijweide PJ,
nol. 2011;209(1):21–32. Burger EH. Pulsating fluid flow increases nitric oxide (NO)
110. Xiao Z, Dallas M, Qiu N, et al. Conditional deletion of synthesis by osteocytes but not periosteal fibroblasts—
Pkd1 in osteocytes disrupts skeletal mechanosensing in correlation with prostaglandin upregulation. Biochem
mice. FASEB J. 2011;25(7):2418 –2432. Biophys Res Commun. 1995;217(2):640 – 648.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


111. Kramer I, Baertschi S, Halleux C, Keller H, Kneissel M. 130. Price C, Zhou X, Li W, Wang L. Real-time measurement of
Mef2c deletion in osteocytes results in increased bone solute transport within the lacunar-canalicular system of
mass. J Bone Miner Res. 2012;27(2):360 –373. mechanically loaded bone: direct evidence for load-in-
112. Nakashima T, Hayashi M, Fukunaga T, et al. Evidence for duced fluid flow. J Bone Miner Res. 2011;26(2):277–285.
osteocyte regulation of bone homeostasis through RANKL 131. Kamel MA, Picconi JL, Lara-Castillo N, Johnson ML. Ac-
expression. Nat Med. 2011;17(10):1231–1234. tivation of ␤-catenin signaling in MLO-Y4 osteocytic cells
113. Xiong J, Onal M, Jilka RL, Weinstein RS, Manolagas SC, versus 2T3 osteoblastic cells by fluid flow shear stress and
O’Brien CA. Matrix-embedded cells control osteoclast for- PGE(2): implications for the study of mechanosensation in
mation. Nat Med. 2011;17(10):1235–1241. bone. Bone. 2010;47(5):872– 881.
114. van der Plas A, Nijweide PJ. Isolation and purification of 132. Kalogeropoulos M, Varanasi SS, Olstad OK, et al. Zic1
osteocytes. J Bone Miner Res. 1992;7(4):389 –396. transcription factor in bone: neural developmental protein
115. Mikuni-Takagaki Y, Suzuki Y, Kawase T, Saito S. Distinct regulates mechanotransduction in osteocytes. FASEB J.
responses of different populations of bone cells to mechan- 2010;24(8):2893–2903.
ical stress. Endocrinology. 1996;137(5):2028 –2035. 133. Lu XL, Huo B, Chiang V, Guo XE. Osteocytic network is
116. Zhao S, Zhang YK, Harris S, Ahuja SS, Bonewald LF. more responsive in calcium signaling than osteoblastic net-
MLO-Y4 osteocyte-like cells support osteoclast formation work under fluid flow. J Bone Miner Res. 2012;27(3):563–
and activation. J Bone Miner Res. 2002;17(11):2068 – 574.
2079. 134. Han Y, Cowin SC, Schaffler MB, Weinbaum S. Mecha-
117. Stern AR, Stern MM, Van Dyke ME, Jahn K, Prideaux M, notransduction and strain amplification in osteocyte cell
Bonewald LF. Isolation and culture of primary osteocytes processes. Proc Natl Acad Sci USA. 2004;101(47):16689 –
from the long bones of skeletally mature and aged mice. 16694.
Biotechniques. 2012;52(6):361–373. 135. Adachi T, Aonuma Y, Tanaka M, Hojo M, Takano-
118. Bonewald LF. Establishment and characterization of an Yamamoto T, Kamioka H. Calcium response in single os-
osteocyte-like cell line, MLO-Y4. J Bone Miner Metab. teocytes to locally applied mechanical stimulus: differences
1999;17(1):61– 65. in cell process and cell body. J Biomech. 2009;42(12):
119. Rosser J, Bonewald LF. Studying osteocyte function using 1989 –1995.
the cell lines MLO-Y4 and MLO-A5. Methods Mol Biol. 136. Nicolella DP, Feng JQ, Moravits DE, et al. Effects of nano-
2012;816:67– 81. mechanical bone tissue properties on bone tissue strain:
120. Kato Y, Windle JJ, Koop BA, Mundy GR, Bonewald LF. implications for osteocyte mechanotransduction. J Mus-
Establishment of an osteocyte-like cell line, MLO-Y4. culoskelet Neuronal Interact. 2008;8(4):330 –331.
J Bone Miner Res. 1997;12(12):2014 –2023. 137. Xiao Z, Zhang S, Mahlios J, et al. Cilia-like structures and
121. Bellows CG, Aubin JE. Determination of numbers of os- polycystin-1 in osteoblasts/osteocytes and associated ab-
teoprogenitors present in isolated fetal rat calvaria cells in normalities in skeletogenesis and Runx2 expression. J Biol
vitro. Dev Biol. 1989;133(1):8 –13. Chem. 2006;281(41):30884 –30895.
122. Aubin JE. Advances in the osteoblast lineage. Biochem Cell 138. Malone AM, Anderson CT, Tummala P, et al. Primary
Biol. 1998;76(6):899 –910. cilia mediate mechanosensing in bone cells by a calcium-
123. Bodine PV, Vernon SK, Komm BS. Establishment and hor- independent mechanism. Proc Natl Acad Sci USA. 2007;
monal regulation of a conditionally transformed preosteo- 104(33):13325–13330.
cytic cell line from adult human bone. Endocrinology. 139. Burra S, Nicolella DP, Francis WL, et al. Dendritic pro-
1996;137(11):4592– 4604. cesses of osteocytes are mechanotransducers that induce
124. Kato Y, Boskey A, Spevak L, Dallas M, Hori M, Bonewald the opening of hemichannels. Proc Natl Acad Sci USA.
LF. Establishment of an osteoid preosteocyte-like cell 2010;107(31):13648 –13653.
MLO-A5 that spontaneously mineralizes in culture. J Bone 140. Kwon RY, Temiyasathit S, Tummala P, Quah CC, Jacobs
Miner Res. 2001;16(9):1622–1633. CR. Primary cilium-dependent mechanosensing is medi-
125. Woo SM, Rosser J, Dusevich V, Kalajzic I, Bonewald LF. ated by adenylyl cyclase 6 and cyclic AMP in bone cells.
Cell line IDG-SW3 replicates osteoblast-to-late-osteocyte FASEB J. 2010;24(8):2859 –2868.
differentiation in vitro and accelerates bone formation in 141. Hoey DA, Kelly DJ, Jacobs CR. A role for the primary
vivo. J Bone Miner Res. 2011;26(11):2634 –2646. cilium in paracrine signaling between mechanically stim-
684 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

ulated osteocytes and mesenchymal stem cells. Biochem production of nitric oxide and prostaglandin E(2) by pri-
Biophys Res Commun. 2011;412(1):182–187. mary bone cells is shear stress dependent. J Biomech. 2001;
142. Qiu N, Cao L, David V, Quarles LD, Xiao Z. Kif3a defi- 34(5):671– 677.
ciency reverses the skeletal abnormalities in Pkd1 deficient 158. Tan SD, Bakker AD, Semeins CM, Kuijpers-Jagtman AM,
mice by restoring the balance between osteogenesis and Klein-Nulend J. Inhibition of osteocyte apoptosis by fluid
adipogenesis. PLoS One. 2010;5(12):e15240. flow is mediated by nitric oxide. Biochem Biophys Res
143. Salter DM, Robb JE, Wright MO. Electrophysiological re- Commun. 2008;369(4):1150 –1154.
sponses of human bone cells to mechanical stimulation: 159. Bakker AD, Silva VC, Krishnan R, et al. Tumor necrosis
evidence for specific integrin function in mechanotrans- factor ␣ and interleukin-1␤ modulate calcium and nitric
duction. J Bone Miner Res. 1997;12(7):1133–1141. oxide signaling in mechanically stimulated osteocytes. Ar-
144. You LD, Weinbaum S, Cowin SC, Schaffler MB. Ultra- thritis Rheum. 2009;60(11):3336 –3345.
structure of the osteocyte process and its pericellular ma- 160. Kamioka H, Miki Y, Sumitani K, et al. Extracellular cal-
trix. Anat Rec A Discov Mol Cell Evol Biol. 2004;278(2): cium causes the release of calcium from intracellular stores

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


505–513. in chick osteocytes. Biochem Biophys Res Commun. 1995;
145. Batra N, Burra S, Siller-Jackson AJ, et al. Mechanical 212(2):692– 696.
stress-activated integrin ␣5␤1 induces opening of connexin 161. Genetos DC, Geist DJ, Liu D, Donahue HJ, Duncan RL.
43 hemichannels. Proc Natl Acad Sci USA. 2012;109(9): Fluid shear-induced ATP secretion mediates prostaglandin
3359 –3364. release in MC3T3–E1 osteoblasts. J Bone Miner Res. 2005;
146. Nicolella DP, Moravits DE, Gale AM, Bonewald LF, Lank- 20(1):41– 49.
ford J. Osteocyte lacunae tissue strain in cortical bone. 162. Cherian PP, Cheng B, Gu S, Sprague E, Bonewald LF, Jiang
J Biomech. 2006;39(9):1735–1743. JX. Effects of mechanical strain on the function of Gap
147. Bonivtch AR, Bonewald LF, Nicolella DP. Tissue strain junctions in osteocytes are mediated through the prosta-
amplification at the osteocyte lacuna: a microstructural glandin EP2 receptor. J Biol Chem. 2003;278(44):43146 –
finite element analysis. J Biomech. 2007;40(10):2199 – 43156.
2206.
163. Cherian PP, Siller-Jackson AJ, Gu S, et al. Mechanical
148. Gu Y, Preston MR, El Haj AJ, Howl JD, Publicover SJ.
strain opens connexin 43 hemichannels in osteocytes: a
Three types of K(⫹) currents in murine osteocyte-like cells
novel mechanism for the release of prostaglandin. Mol Biol
(MLO-Y4). Bone. 2001;28(1):29 –37.
Cell. 2005;16(7):3100 –3106.
149. Shao Y, Alicknavitch M, Farach-Carson MC. Expression
164. Qiang YW, Chen Y, Brown N, et al. Characterization of
of voltage sensitive calcium channel (VSCC) L-type Cav1.2
Wnt/␤-catenin signalling in osteoclasts in multiple my-
(␣1C) and T-type Cav3.2 (␣1H) subunits during mouse
eloma. Br J Haematol. 2010;148(5):726 –738.
bone development. Dev Dyn. 2005;234(1):54 – 62.
165. Tang LY, Cullen DM, Yee JA, Jee WS, Kimmel DB. Pros-
150. Gu Y, Preston MR, Magnay J, El Haj AJ, Publicover SJ.
taglandin E2 increases the skeletal response to mechanical
Hormonally-regulated expression of voltage-operated
loading. J Bone Miner Res. 1997;12(2):276 –282.
Ca(2⫹) channels in osteocytic (MLO-Y4) cells. Biochem
166. Pead MJ, Lanyon LE. Indomethacin modulation of load-
Biophys Res Commun. 2001;282(2):536 –542.
151. Thompson WR, Majid AS, Czymmek KJ, et al. Association related stimulation of new bone formation in vivo. Calcif
of the ␣(2)␦(1) subunit with Ca (v) 3.2 enhances membrane Tissue Int. 1989;45(1):34 – 40.
expression and regulates mechanically induced ATP re- 167. Grimston SK, Brodt MD, Silva MJ, Civitelli R. Attenuated
lease in MLO-Y4 osteocytes. J Bone Miner Res. 2011; response to in vivo mechanical loading in mice with con-
26(9):2125–2139. ditional osteoblast ablation of the connexin43 gene (Gja1).
152. Forwood MR. Inducible cyclo-oxygenase (COX-2) medi- J Bone Miner Res. 2008;23(6):879 – 886.
ates the induction of bone formation by mechanical load- 168. Zhang Y, Paul EM, Sathyendra V, et al. Enhanced oste-
ing in vivo. J Bone Miner Res. 1996;11(11):1688 –1693. oclastic resorption and responsiveness to mechanical load
153. Watanuki M, Sakai A, Sakata T, et al. Role of inducible in gap junction deficient bone. PLoS One. 2011;6(8):
nitric oxide synthase in skeletal adaptation to acute in- e23516.
creases in mechanical loading. J Bone Miner Res. 2002; 169. Loiselle AE, Jiang JX, Donahue HJ. Gap junction and he-
17(6):1015–1025. michannel functions in osteocytes. Bone. 2013;54(2):205–
154. Kunnel JG, Igarashi K, Gilbert JL, Stern PH. Bone anabolic 212.
responses to mechanical load in vitro involve COX-2 and 170. Xia X, Batra N, Shi Q, Bonewald LF, Sprague E, Jiang JX.
constitutive NOS. Connect Tissue Res. 2004;45(1):40 – Prostaglandin promotion of osteocyte gap junction func-
49. tion through transcriptional regulation of connexin 43 by
155. Li J, Liu D, Ke HZ, Duncan RL, Turner CH. The P2X7 glycogen synthase kinase 3/␤-catenin signaling. Mol Cell
nucleotide receptor mediates skeletal mechanotransduc- Biol. 2010;30(1):206 –219.
tion. J Biol Chem. 2005;280(52):42952– 42959. 171. Kitase Y, Barragan L, Qing H, et al. Mechanical induction
156. Wang N, Rumney RM, Yang L, et al. The P2Y13 receptor of PGE2 in osteocytes blocks glucocorticoid-induced ap-
regulates extracellular ATP metabolism and the osteogenic optosis through both the ␤-catenin and PKA pathways.
response to mechanical loading [published online ahead of J Bone Miner Res. 2010;25(12):2657–2668.
print January 29, 2013]. J Bone Miner Res. doi:10.1002/ 172. Sawakami K, Robling AG, Ali M, et al. The Wnt co-re-
jbmr.1877. ceptor LRP5 is essential for skeletal mechanotransduction
157. Bakker AD, Soejima K, Klein-Nulend J, Burger EH. The but not for anabolic bone response to parathyroid hor-
doi: 10.1210/er.2012-1026 edrv.endojournals.org 685

mone treatment. J Biol Chem. 2006;281(33):23698 – 188. Raisz LG, Fall PM, Gabbitas BY, McCarthy TL, Kream
23711. BE, Canalis E. Effects of prostaglandin E2 on bone for-
173. Lin C, Jiang X, Dai Z, et al. Sclerostin mediates bone re- mation in cultured fetal rat calvariae: role of insulin-like
sponse to mechanical unloading through antagonizing growth factor-I. Endocrinology. 1993;133(4):1504 –
Wnt/␤-catenin signaling. J Bone Miner Res. 2009;24(10): 1510.
1651–1661. 189. Nagata T, Kaho K, Nishikawa S, Shinohara H, Wakano Y,
174. Javaheri B, Dallas M, Zhao H, Bonewald L, Johnson M. Ishida H. Effect of prostaglandin E2 on mineralization of
b-Catenin haploinsufficiency in osteocytes abolishes the bone nodules formed by fetal rat calvarial cells. Calcif Tis-
osteogenic effect of mechanical loading in vivo. J Bone sue Int. 1994;55(6):451– 457.
Miner Res. 2011;26(suppl 1):S24. 190. Raisz LG, Kream BE. Regulation of bone formation (sec-
175. Lee K, Jessop H, Suswillo R, Zaman G, Lanyon L. Endo- ond of two parts). N Engl J Med. 1983;309(2):83– 89.
crinology: bone adaptation requires oestrogen receptor-␣. 191. Jee WS, Ueno K, Deng YP, Woodbury DM. The effects of
Nature. 2003;424(6947):389. prostaglandin E2 in growing rats: increased metaphyseal

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


176. Zaman G, Jessop HL, Muzylak M, et al. Osteocytes use hard tissue and cortico-endosteal bone formation. Calcif
estrogen receptor ␣ to respond to strain but their ER␣ Tissue Int. 1985;37(2):148 –157.
content is regulated by estrogen. J Bone Miner Res. 2006; 192. Gong Y, Slee RB, Fukai N, et al. LDL receptor-related
21(8):1297–1306. protein 5 (LRP5) affects bone accrual and eye develop-
177. Yang W, Lu Y, Kalajzic I, et al. Dentin matrix protein 1 ment. Cell. 2001;107(4):513–523.
gene cis-regulation: use in osteocytes to characterize local 193. Kato M, Patel MS, Levasseur R, et al. Cbfa 1-independent
responses to mechanical loading in vitro and in vivo. J Biol decrease in osteoblast proliferation, osteopenia, and per-
Chem. 2005;280(21):20680 –20690. sistent embryonic eye vascularization in mice deficient in
178. Gluhak-Heinrich J, Ye L, Bonewald LF, et al. Mechanical Lrp5, a Wnt coreceptor. J Cell Biol. 2002;157:303–314.
loading stimulates dentin matrix protein 1 (DMP1) expres- 194. Zhang Y, Wang Y, Li X, et al. The LRP5 high-bone-mass
sion in osteocytes in vivo. J Bone Miner Res. 2003;18(5): G171V mutation disrupts LRP5 interaction with Mesd.
Mol Cell Biol. 2004;24(11):4677– 4684.
807– 817.
195. Li J, Sarosi I, Cattley RC, et al. Dkk1-mediated inhibition
179. Gluhak-Heinrich J, Pavlin D, Yang W, Macdougall M,
of Wnt signaling in bone results in osteopenia. Bone. 2006;
Harris SE. MEPE expression in osteocytes during orth-
39(4):754 –766.
odontic tooth movement. Arch Oral Biol. 2007;52(7):
196. Heiland GR, Zwerina K, Baum W, et al. Neutralisation of
684 – 690.
Dkk-1 protects from systemic bone loss during inflamma-
180. Vezeridis PS, Semeins CM, Chen Q, Klein-Nulend J. Os-
tion and reduces sclerostin expression. Ann Rheum Dis.
teocytes subjected to pulsating fluid flow regulate osteo-
2010;69(12):2152–2159.
blast proliferation and differentiation. Biochem Biophys
197. Bodine PV, Zhao W, Kharode YP, et al. The Wnt antag-
Res Commun. 2006;348(3):1082–1088.
onist secreted frizzled-related protein-1 is a negative reg-
181. Heino TJ, Hentunen TA, Vaananen HK. Conditioned me-
ulator of trabecular bone formation in adult mice. Mol
dium from osteocytes stimulates the proliferation of bone
Endocrinol. 2004;18(5):1222–1237.
marrow mesenchymal stem cells and their differentiation 198. Bodine PV, Billiard J, Moran RA, et al. The Wnt antagonist
into osteoblasts. Exp Cell Res. 2004;294(2):458 – 468. secreted frizzled-related protein-1 controls osteoblast and
182. Raheja LF, Genetos DC, Yellowley CE. Hypoxic osteo- osteocyte apoptosis. J Cell Biochem. 2005;96(6):1212–
cytes recruit human MSCs through an OPN/CD44-medi- 1230.
ated pathway. Biochem Biophys Res Commun. 2008; 199. Tanaka K, Yamaguchi Y, Hakeda Y. Isolated chick osteo-
366(4):1061–1066. cytes stimulate formation and bone-resorbing activity of
183. Zaman G, Pitsillides AA, Rawlinson SC, et al. Mechanical osteoclast-like cells. J Bone Miner Metab. 1995;13:61–70.
strain stimulates nitric oxide production by rapid activa- 200. Kartsogiannis V, Zhou H, Horwood NJ, et al. Localization
tion of endothelial nitric oxide synthase in osteocytes. of RANKL (receptor activator of NF ␬ B ligand) mRNA
J Bone Miner Res. 1999;14(7):1123–1131. and protein in skeletal and extraskeletal tissues. Bone.
184. Papanicolaou SE, Phipps RJ, Fyhrie DP, Genetos DC. 1999;25(5):525–534.
Modulation of sclerostin expression by mechanical load- 201. Silvestrini G, Ballanti P, Patacchioli F, et al. Detection of
ing and bone morphogenetic proteins in osteogenic cells. osteoprotegerin (OPG) and its ligand (RANKL) mRNA
Biorheology. 2009;46(5):389 –399. and protein in femur and tibia of the rat. J Mol Histol.
185. Basso N, Heersche JN. Effects of hind limb unloading and 2005;36(1–2):59 – 67.
reloading on nitric oxide synthase expression and apopto- 202. Kurata K, Heino TJ, Higaki H, Vaananen HK. Bone mar-
sis of osteocytes and chondrocytes. Bone. 2006;39(4):807– row cell differentiation induced by mechanically damaged
814. osteocytes in 3D gel-embedded culture. J Bone Miner Res.
186. Caballero-Alias AM, Loveridge N, Lyon A, Das-Gupta V, 2006;21(4):616 – 625.
Pitsillides A, Reeve J. NOS isoforms in adult human os- 203. Kulkarni RN, Bakker AD, Everts V, Klein-Nulend J. In-
teocytes: multiple pathways of NO regulation? Calcif Tis- hibition of osteoclastogenesis by mechanically loaded os-
sue Int. 2004;75(1):78 – 84. teocytes: involvement of MEPE. Calcif Tissue Int. 2010;
187. Blackwell KA, Raisz LG, Pilbeam CC. Prostaglandins in 87(5):461– 468.
bone: bad cop, good cop? Trends Endocrinol Metab. 2010; 204. Ikeda T, Utsuyama M, Hirokawa K. Expression profiles of
21(5):294 –301. receptor activator of nuclear factor ␬B ligand, receptor
686 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

activator of nuclear factor ␬B, and osteoprotegerin mes- mechanotransduction pathway. Am J Physiol. 1999;277:
senger RNA in aged and ovariectomized rat bones. J Bone C598 –C602.
Miner Res. 2001;16(8):1416 –1425. 221. Weinstein RS, Nicholas RW, Manolagas SC. Apoptosis of
205. Gu G, Mulari M, Peng Z, Hentunen TA, Vaananen HK. osteocytes in glucocorticoid-induced osteonecrosis of the
Death of osteocytes turns off the inhibition of osteoclasts hip. J Clin Endocrinol Metab. 2000;85(8):2907–2912.
and triggers local bone resorption. Biochem Biophys Res 222. Weinstein RS, Jilka RL, Parfitt AM, Manolagas SC. Inhi-
Commun. 2005;335(4):1095–1101. bition of osteoblastogenesis and promotion of apoptosis of
206. Heino TJ, Kurata K, Higaki H, Vaananen HK. Evidence osteoblasts and osteocytes by glucocorticoids. Potential
for the role of osteocytes in the initiation of targeted re- mechanisms of their deleterious effects on bone. J Clin In-
modeling. Technol Health Care. 2009;17(1):49 –56. vest. 1998;102(2):274 –282.
207. Yang J, Shah R, Robling AG, et al. HMGB1 is a bone-active 223. O’Brien CA, Jia D, Plotkin LI, et al. Glucocorticoids act
cytokine. J Cell Physiol. 2008;214(3):730 –739. directly on osteoblasts and osteocytes to induce their ap-
208. Kogianni G, Mann V, Noble BS. Apoptotic bodies convey optosis and reduce bone formation and strength. Endocri-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


activity capable of initiating osteoclastogenesis and local- nology. 2004;145(4):1835–1841.
ized bone destruction. J Bone Miner Res. 2008;23(6):915– 224. Plotkin LI, Manolagas SC, Bellido T. Glucocorticoids in-
927. duce osteocyte apoptosis by blocking focal adhesion ki-
209. Tan SD, de Vries TJ, Kuijpers-Jagtman AM, Semeins CM, nase-mediated survival. Evidence for inside-out signaling
Everts V, Klein-Nulend J. Osteocytes subjected to fluid leading to anoikis. J Biol Chem. 2007;282(33):24120 –
flow inhibit osteoclast formation and bone resorption. 24130.
Bone. 2007;41(5):745–751. 225. Pacifici R, Brown C, Puscheck E, et al. Effect of surgical
210. Zhao Q, Brauer PR, Xiao L, McGuire MH, Yee JA. Ex- menopause and estrogen replacement on cytokine release
pression of parathyroid hormone-related peptide (PthrP) from human blood mononuclear cells. Proc Natl Acad Sci
and its receptor (PTH1R) during the histogenesis of carti- USA. 1991;88(12):5134 –5138.
lage and bone in the chicken mandibular process. J Anat. 226. Al-Dujaili SA, Lau E, Al-Dujaili H, Tsang K, Guenther A,
2002;201(2):137–151.
You L. Apoptotic osteocytes regulate osteoclast precursor
211. Lupp A, Klenk C, Rocken C, Evert M, Mawrin C, Schulz
recruitment and differentiation in vitro. J Cell Biochem.
S. Immunohistochemical identification of the PTHR1
2011;112(9):2412–2423.
parathyroid hormone receptor in normal and neoplastic
227. Mulcahy LE, Taylor D, Lee TC, Duffy GP. RANKL and
human tissues. Eur J Endocrinol. 2010;162(5):979 –986.
OPG activity is regulated by injury size in networks of
212. Bellido T, Ali AA, Gubrij I, et al. Chronic elevation of
osteocyte-like cells. Bone. 2011;48(2):182–188.
parathyroid hormone in mice reduces expression of
228. Mabilleau G, Mieczkowska A, Edmonds ME. Thiazoli-
sclerostin by osteocytes: a novel mechanism for hormonal
dinediones induce osteocyte apoptosis and increase
control of osteoblastogenesis. Endocrinology. 2005;
sclerostin expression. Diabet Med. 2010;27(8):925–932.
146(11):4577– 4583.
229. Maurel DB, Jaffre C, Rochefort GY, et al. Low bone ac-
213. Keller H, Kneissel M. SOST is a target gene for PTH in
crual is associated with osteocyte apoptosis in alcohol-in-
bone. Bone. 2005;37(2):148 –158.
214. O’Brien CA, Plotkin LI, Galli C, et al. Control of bone mass duced osteopenia. Bone. 2011;49(3):543–552.
and remodeling by PTH receptor signaling in osteocytes. 230. Shandala T, Shen Ng Y, Hopwood B, Yip YC, Foster BK,
PLoS One. 2008;3(8):e2942. Xian CJ. The role of osteocyte apoptosis in cancer chemo-
215. Qing H, Ardeshirpour L, Pajevic PD, et al. Demonstration therapy-induced bone loss. J Cell Physiol. 2012;227(7):
of osteocytic perilacunar/canalicular remodeling in mice 2889 –2897.
during lactation. J Bone Miner Res. 2012;27(5):1018 – 231. Bonewald L. Osteocytes. In: Marcus R, Nelson D, Rosen
1029. C, eds. Osteoporosis. Vol 1. 3rd ed. Burlington, MA:
216. Verborgt O, Gibson GJ, Schaffler MB. Loss of osteocyte Elsevier; 2008:169 –190.
integrity in association with microdamage and bone re- 232. Weinstein RS, Jilka RL, Almeida M, Roberson PK, Mano-
modeling after fatigue in vivo. J Bone Miner Res. 2000; lagas SC. Intermittent parathyroid hormone administra-
15(1):60 – 67. tion counteracts the adverse effects of glucocorticoids on
217. Verborgt O, Tatton NA, Majeska RJ, Schaffler MB. Spa- osteoblast and osteocyte viability, bone formation, and
tial distribution of Bax and Bcl-2 in osteocytes after bone strength in mice. Endocrinology. 2010;151(6):2641–
fatigue: complementary roles in bone remodeling regula- 2649.
tion? J Bone Miner Res. 2002;17(5):907–914. 233. Yajima A, Inaba M, Tominaga Y, Nishizawa Y, Ikeda K,
218. Kennedy OD, Herman BC, Laudier DM, Majeska RJ, Sun Ito A. Increased osteocyte death and mineralization inside
HB, Schaffler MB. Activation of resorption in fatigue- bone after parathyroidectomy in patients with secondary
loaded bone involves both apoptosis and active pro-oste- hyperparathyroidism. J Bone Miner Res. 2010;25(11):
oclastogenic signaling by distinct osteocyte populations. 2374 –2381.
Bone. 2012;50(5):1115–1122. 234. Kousteni S, Bellido T, Plotkin LI, et al. Nongenotropic,
219. Tomkinson A, Reeve J, Shaw RW, Noble BS. The death of sex-nonspecific signaling through the estrogen or andro-
osteocytes via apoptosis accompanies estrogen withdrawal gen receptors: dissociation from transcriptional activity.
in human bone. J Clin Endocrinol Metab. 1997;82(9): Cell. 2001;104(5):719 –730.
3128 –3135. 235. Aguirre JI, Plotkin LI, Gortazar AR, et al. A novel ligand-
220. Dodd JS, Raleigh JA, Gross TS. Osteocyte hypoxia: a novel independent function of the estrogen receptor is essential
doi: 10.1210/er.2012-1026 edrv.endojournals.org 687

for osteocyte and osteoblast mechanotransduction. J Biol conditions and under the influence of parathyroid extract
Chem. 2007;282(35):25501–25508. [in French]. Schweiz Med Wochenschr. 1968;98(19):717–
236. Weinstein RS, O’Brien CA, Almeida M, et al. Osteopro- 720.
tegerin prevents glucocorticoid-induced osteocyte apopto- 253. Baud CA. Submicroscopic structure and functional aspects
sis in mice. Endocrinology. 2011;152(9):3323–3331. of the osteocyte. Clin Orthop Relat Res. 1968;56:227–
237. Bellido T. Antagonistic interplay between mechanical 236.
forces and glucocorticoids in bone: a tale of kinases. J Cell 254. Belanger LF, Robichon J. Parathormone-induced osteoly-
Biochem. 2010;111(1):1– 6. sis in dogs. A microradiographic and alpharadiographic
238. Plotkin LI, Lezcano V, Thostenson J, Weinstein RS, Mano- survey. J Bone Joint Surg Am. 1964;46:1008 –1012.
lagas SC, Bellido T. Connexin 43 is required for the anti- 255. Iagodovskii VS, Triftanidi LA, Gorokhova GP. Effect of
apoptotic effect of bisphosphonates on osteocytes and space flight on rat skeletal bones (an optical light and elec-
osteoblasts in vivo. J Bone Miner Res. 2008;23(11): tron microscopic study) [in Russian]. Kosm Biol Aviakosm
Med. 1977;11(1):14 –20.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


1712–1721.
239. Bivi N, Condon KW, Allen MR, et al. Cell autonomous 256. Haller AC, Zimny ML. Effects of hibernation on interra-
requirement of connexin 43 for osteocyte survival: conse- dicular alveolar bone. J Dent Res. 1977;56(12):1552–
quences for endocortical resorption and periosteal bone 1557.
formation. J Bone Miner Res. 2012;27(2):374 –389. 257. Alcobendas M, Baud CA, Castanet J. Structural changes of
240. Follet H, Li J, Phipps RJ, Hui S, Condon K, Burr DB. the periosteocytic area in Vipera aspis (L.) (Ophidia, Vi-
Risedronate and alendronate suppress osteocyte apoptosis peridae) bone tissue in various physiological conditions.
following cyclic fatigue loading. Bone. 2007;40(4):1172– Calcif Tissue Int. 1991;49(1):53–57.
1177. 258. Heller-Steinberg M. Ground substance, bone salts, and cel-
241. Plotkin LI, Weinstein RS, Parfitt AM, Roberson PK, lular activity in bone formation and destruction. Am J
Manolagas SC, Bellido T. Prevention of osteocyte and os- Anat. 1951;89(3):347–379.
teoblast apoptosis by bisphosphonates and calcitonin. 259. Ruth EB. Basophilic islands in osseous tissue and their
relation to resorption. Anat Rec. 1961;140:307–320.
J Clin Invest. 1999;104(10):1363–1374.
260. Wergedal JE, Baylink DJ. Distribution of acid and alkaline
242. Plotkin LI, Mathov I, Aguirre JI, Parfitt AM, Manolagas
phosphatase activity in undemineralized sections of the rat
SC, Bellido T. Mechanical stimulation prevents osteocyte
tibial diaphysis. J Histochem Cytochem. 1969;17(12):
apoptosis: requirement of integrins, Src kinases, and ERKs.
799 – 806.
Am J Physiol Cell Physiol. 2005;289(3):C633–C643.
261. Nakano Y, Toyosawa S, Takano Y. Eccentric localization
243. Emerton KB, Hu B, Woo AA, et al. Osteocyte apoptosis
of osteocytes expressing enzymatic activities, protein, and
and control of bone resorption following ovariectomy in
mRNA signals for type 5 tartrate-resistant acid phospha-
mice. Bone. 2010;46(3):577–583.
tase (TRAP). J Histochem Cytochem. 2004;52(11):1475–
244. Xia X, Kar R, Gluhak-Heinrich J, et al. Glucocorticoid-
1482.
induced autophagy in osteocytes. J Bone Miner Res. 2010;
262. Baylink DJ, Wergedal JE. Bone formation by osteocytes.
25(11):2479 –2488.
Am J Physiol. 1971;221(3):669 – 678.
245. Jia J, Yao W, Guan M, et al. Glucocorticoid dose deter- 263. Zambonin-Zallone A, Teti A, Primavera MV, Pace G. Ma-
mines osteocyte cell fate. FASEB J. 2011;25(10):3366 – ture osteocytes behaviour in a repletion period: the occur-
3376. rence of osteoplastic activity. Basic Appl Histochem. 1983;
246. Zhao H, Xiong J, Onal M, et al. Osteocyte autophagy 27(3):191–204.
declines with age in mice and suppression of autophagy 264. Yamashita T, Yoshioka M, Itoh N. Identification of a
decreases bone mass. J Bone Miner Res. 2011;27(Suppl1): novel fibroblast growth factor, FGF-23, preferentially ex-
S13. pressed in the ventrolateral thalamic nucleus of the brain.
247. Marotti G, Ferretti M, Remaggi F, Palumbo C. Quantita- Biochem Biophys Res Commun. 2000;277(2):494 – 498.
tive evaluation on osteocyte canalicular density in human 265. Shimada T, Kakitani M, Yamazaki Y, et al. Targeted ab-
secondary osteons. Bone. 1995;16(1):125–128. lation of Fgf23 demonstrates an essential physiological
248. Belanger LF, Belanger C, Semba T. Technical approaches role of FGF23 in phosphate and vitamin D metabolism.
leading to the concept of osteocytic osteolysis. Clin Orthop J Clin Invest. 2004;113(4):561–568.
Relat Res. 1967;54:187–196. 266. Liu S, Lu Y, Xie Y, et al. Elevated levels of FGF23 in dentin
249. Recklinghausen FV. Untersuchungen uber Rachitis und matrix protein 1 (DMP1) null mice potentially explain phe-
Osteomalacia. Jena, Germany: Gustav Fischer; 1910. notypic similarities to Hyp mice. J Bone Miner Res. 2006;
250. Bonucci E, Gherardi G, Mioni G, et al. Clinico-morpho- 21(Suppl1):S51.
logical correlations in uremic osteodystrophy of patients 267. Riminucci M, Collins MT, Fedarko NS, et al. FGF-23 in
with conservative and hemodialytic treatment with special fibrous dysplasia of bone and its relationship to renal phos-
regard to the ultrastructure [in Italian]. Minerva Nefrol. phate wasting. J Clin Invest. 2003;112(5):683– 692.
1975;22(2–3):99 –108. 268. Urakawa I, Yamazaki Y, Shimada T, et al. Klotho converts
251. Bonucci E, Gherardi G. Osteocyte ultrastructure in renal canonical FGF receptor into a specific receptor for FGF23.
osteodystrophy. Virchows Arch A Pathol Anat Histol. Nature. 2006;444(7120):770 –774.
1977;373(3):213–231. 269. Gattineni J, Bates C, Twombley K, et al. FGF23 decreases
252. Baud CA. Structure and functions of osteocytes in normal renal NaPi-2a and NaPi-2c expression and induces hy-
688 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

pophosphatemia in vivo predominantly via FGF receptor but not degradation by PHEX. J Biol Chem. 2003;278(39):
1. Am J Physiol Renal Physiol. 2009;297(2):F282–F291. 37419 –37426.
270. Larsson T, Marsell R, Schipani E, et al. Transgenic mice 285. Martin A, Liu S, David V, et al. Bone proteins PHEX and
expressing fibroblast growth factor 23 under the control of DMP1 regulate fibroblastic growth factor Fgf23 expres-
the ␣1(I) collagen promoter exhibit growth retardation, sion in osteocytes through a common pathway involving
osteomalacia, and disturbed phosphate homeostasis. En- FGF receptor (FGFR) signaling. FASEB J. 2011;25(8):
docrinology. 2004;145(7):3087–3094. 2551–2562.
271. Miyamoto K, Ito M, Kuwahata M, Kato S, Segawa H. 286. Martin A, David V, Laurence JS, et al. Degradation of
Inhibition of intestinal sodium-dependent inorganic phos- MEPE, DMP1, and release of SIBLING ASARM-peptides
phate transport by fibroblast growth factor 23. Ther Apher (minhibins): ASARM-peptide (s) are directly responsible
Dial. 2005;9(4):331–335. for defective mineralization in HYP. Endocrinology. 2008;
272. Liu S, Zhou J, Tang W, Jiang X, Rowe DW, Quarles LD. 149(4):1757–1772.
Pathogenic role of Fgf23 in Hyp mice. Am J Physiol En- 287. Gowen LC, Petersen DN, Mansolf AL, et al. Targeted dis-

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


docrinol Metab. 2006;291(1):E38 –E49. ruption of the osteoblast/osteocyte factor 45 gene (OF45)
273. Perwad F, Azam N, Zhang MY, Yamashita T, Tenenhouse results in increased bone formation and bone mass. J Biol
HS, Portale AA. Dietary and serum phosphorus regulate Chem. 2003;278(3):1998 –2007.
fibroblast growth factor 23 expression and 1,25-dihy- 288. Rowe PS, Garrett IR, Schwarz PM, et al. Surface plasmon
droxyvitamin D metabolism in mice. Endocrinology. resonance (SPR) confirms that MEPE binds to PHEX via
2005;146(12):5358 –5364. the MEPE-ASARM motif: a model for impaired mineral-
274. Burnett SM, Gunawardene SC, Bringhurst FR, Juppner H, ization in X-linked rickets (HYP). Bone. 2005;36(1):33–
Lee H, Finkelstein JS. Regulation of C-terminal and intact 46.
FGF-23 by dietary phosphate in men and women. J Bone 289. Guo R, Rowe PS, Liu S, Simpson LG, Xiao ZS, Quarles LD.
Miner Res. 2006;21(8):1187–1196. Inhibition of MEPE cleavage by Phex. Biochem Biophys
275. Lavi-Moshayoff V, Wasserman G, Meir T, Silver J, Naveh- Res Commun. 2002;297(1):38 – 45.
Many T. PTH increases FGF23 gene expression and me-
290. Jain A, Fedarko NS, Collins MT, et al. Serum levels of
diates the high-FGF23 levels of experimental kidney fail-
matrix extracellular phosphoglycoprotein (MEPE) in nor-
ure: a bone parathyroid feedback loop. Am J Physiol Renal
mal humans correlate with serum phosphorus, parathy-
Physiol. 2010;299(4):F882–F889.
roid hormone and bone mineral density. J Clin Endocrinol
276. Rhee Y, Bivi N, Farrow E, et al. Parathyroid hormone
Metab. 2004;89(8):4158 – 4161.
receptor signaling in osteocytes increases the expression of
291. Dobbie H, Unwin RJ, Faria NJ, Shirley DG. Matrix ex-
fibroblast growth factor-23 in vitro and in vivo. Bone.
tracellular phosphoglycoprotein causes phosphaturia in
2011;49(4):636 – 643.
rats by inhibiting tubular phosphate reabsorption. Neph-
277. Krajisnik T, Bjorklund P, Marsell R, et al. Fibroblast
rol Dial Transplant. 2008;23(2):730 –733.
growth factor-23 regulates parathyroid hormone and 1␣-
292. Marks J, Churchill LJ, Debnam ES, Unwin RJ. Matrix
hydroxylase expression in cultured bovine parathyroid
extracellular phosphoglycoprotein inhibits phosphate
cells. J Endocrinol. 2007;195(1):125–131.
278. Ben-Dov IZ, Galitzer H, Lavi-Moshayoff V, et al. The transport. J Am Soc Nephrol. 2008;19(12):2313–2320.
parathyroid is a target organ for FGF23 in rats. J Clin 293. David V, Martin A, Hedge AM, Drezner MK, Rowe PS.
Invest. 2007;117(12):4003– 4008. ASARM peptides: PHEX-dependent and -independent
279. Mirza MA, Larsson A, Lind L, Larsson TE. Circulating regulation of serum phosphate. Am J Physiol Renal
fibroblast growth factor-23 is associated with vascular Physiol. 2011;300(3):F783–F791.
dysfunction in the community. Atherosclerosis. 2009; 294. Bresler D, Bruder J, Mohnike K, Fraser WD, Rowe PS.
205(2):385–390. Serum MEPE-ASARM-peptides are elevated in X-linked
280. Faul C, Amaral AP, Oskouei B, et al. FGF23 induces left rickets (HYP): implications for phosphaturia and rickets.
ventricular hypertrophy. J Clin Invest. 2011;121(11): J Endocrinol. 2004;183(3):R1–R9.
4393– 4408. 295. David V, Martin A, Hedge AM, Rowe PS. Matrix extra-
281. Desjardins L, Liabeuf S, Renard C, et al. FGF23 is inde- cellular phosphoglycoprotein (MEPE) is a new bone renal
pendently associated with vascular calcification but not hormone and vascularization modulator. Endocrinology.
bone mineral density in patients at various CKD stages. 2009;150(9):4012– 4023.
Osteoporos Int. 2012;23(7):2017–2025. 296. Hamrick MW. A role for myokines in muscle-bone inter-
282. Mirza MA, Alsio J, Hammarstedt A, et al. Circulating fi- actions. Exerc Sport Sci Rev. 2011;39(1):43– 47.
broblast growth factor-23 is associated with fat mass and 297. Pedersen BK. Muscles and their myokines. J Exp Biol.
dyslipidemia in two independent cohorts of elderly indi- 2011;214:337–346.
viduals. Arterioscler Thromb Vasc Biol. 2011;31(1):219 – 298. Broholm C, Laye MJ, Brandt C, et al. LIF is a contraction-
227. induced myokine stimulating human myocyte prolifera-
283. Rowe PS, Oudet CL, Francis F, et al. Distribution of mu- tion. J Appl Physiol. 2011;111(1):251–259.
tations in the PEX gene in families with X-linked hy- 299. Hamrick MW, Shi X, Zhang W, et al. Loss of myostatin
pophosphataemic rickets (HYP). Hum Mol Genet. 1997; (GDF8) function increases osteogenic differentiation of
6(4):539 –549. bone marrow-derived mesenchymal stem cells but the os-
284. Liu S, Guo R, Simpson LG, Xiao ZS, Burnham CE, Quarles teogenic effect is ablated with unloading. Bone. 2007;
LD. Regulation of fibroblastic growth factor 23 expression 40(6):1544 –1553.
doi: 10.1210/er.2012-1026 edrv.endojournals.org 689

300. Hamrick MW, McNeil PL, Patterson SL. Role of muscle- thyroid hormone (PTH)-induced bone gain is blunted in
derived growth factors in bone formation. J Musculoskelet SOST overexpressing and deficient mice. J Bone Miner
Neuronal Interact. 2010;10(1):64 –70. Res. 2010;25(2):178 –189.
301. Jähn K, Lara-Castillo N, Brotto L, et al. Skeletal muscle 317. Drake MT, Srinivasan B, Modder UI, et al. Effects of para-
secreted factors prevent glucocorticoid-induced osteocyte thyroid hormone treatment on circulating sclerostin levels
apoptosis through activation of ␤-catenin. Eur Cell Mater. in postmenopausal women. J Clin Endocrinol Metab.
2012;24:197–209. 2010;95(11):5056 –5062.
302. Lara N, Jaehn K, Romero-Suarez S, et al. A muscle derived 318. Mirza FS, Padhi ID, Raisz LG, Lorenzo JA. Serum scleros-
soluble factor activates PI3K/␤-catenin signaling in osteo- tin levels negatively correlate with parathyroid hormone
cytes. J Bone Miner Res. 2011;26(suppl 1):S242. levels and free estrogen index in postmenopausal women.
303. Mo C, Romero-Suarez S, Bonewald L, Johnson M, Brotto J Clin Endocrinol Metab. 2010;95(4):1991–1997.
M. Prostaglandin E2: from clinical applications to its po- 319. Ominsky MS, Vlasseros F, Jolette J, et al. Two doses of
tential role in bone-muscle crosstalk and myogenic differ- sclerostin antibody in cynomolgus monkeys increases bone

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020


entiation. Recent Pat Biotechnol. 2012;6(3):223–229. formation, bone mineral density, and bone strength. J Bone
304. Yellowley CE, Li Z, Zhou Z, Jacobs CR, Donahue HJ. Miner Res. 2010;25(5):948 –959.
Functional gap junctions between osteocytic and osteo- 320. Tian X, Setterberg RB, Li X, Paszty C, Ke HZ, Jee WS.
blastic cells. J Bone Miner Res. 2000;15(2):209 –217. Treatment with a sclerostin antibody increases cancellous
305. Qiu S, Rao DS, Palnitkar S, Parfitt AM. Reduced iliac can- bone formation and bone mass regardless of marrow com-
cellous osteocyte density in patients with osteoporotic ver- position in adult female rats. Bone. 2010;47(3):529 –533.
tebral fracture. J Bone Miner Res. 2003;18(9):1657–1663. 321. Li X, Warmington KS, Niu QT, et al. Inhibition of scleros-
306. Loveridge N, Fletcher S, Power J, et al. Patterns of osteo- tin by monoclonal antibody increases bone formation,
cytic endothelial nitric oxide synthase expression in the bone mass, and bone strength in aged male rats. J Bone
femoral neck cortex: differences between cases of intra- Miner Res. 2010;25(12):2647–2656.
capsular hip fracture and controls. Bone. 2002;30(6):866 – 322. Marenzana M, Greenslade K, Eddleston A, et al. Sclerostin an-
871. tibody treatment enhances bone strength but does not prevent
307. Caballero-Alias AM, Loveridge N, Pitsillides A, et al. Os- growth retardation in young mice treated with dexamethasone.
teocytic expression of constitutive NO synthase isoforms Arthritis Rheum. 2011;63(8):2385–2395.
in the femoral neck cortex: a case-control study of intra- 323. Rudmann DG, Page TJ, Vahle JL, et al. Rat-specific decreases in
capsular hip fracture. J Bone Miner Res. 2005;20(2):268 – platelet count caused by a humanized monoclonal antibody
273. against sclerostin. Toxicol Sci. 2012;125(2):586–594.
308. Calder JD, Buttery L, Revell PA, Pearse M, Polak JM. Apo- 324. ADHR Consortium. Autosomal dominant hypophospha-
ptosis—a significant cause of bone cell death in osteone- taemic rickets is associated with mutations in FGF23. Nat
crosis of the femoral head. J Bone Joint Surg Br. 2004; Genet. 2000;26(3):345–348.
86(8):1209 –1213. 325. Larsson T, Nisbeth U, Ljunggren O, Juppner H, Jonsson
309. Humphreys S, Spencer JD, Tighe JR, Cumming RR. The KB. Circulating concentration of FGF-23 increases as renal
femoral head in osteonecrosis. A quantitative study of os- function declines in patients with chronic kidney disease,
teocyte population. J Bone Joint Surg Br. 1989;71(2):205– but does not change in response to variation in phosphate
208. intake in healthy volunteers. Kidney Int. 2003;64(6):
310. Perrotta I, Cristofaro MG, Amantea M, et al. Jaw osteonecrosis 2272–2279.
in patients treated with bisphosphonates: an ultrastructural 326. Imanishi Y, Inaba M, Nakatsuka K, et al. FGF-23 in pa-
study. Ultrastruct Pathol. 2010;34(4):207–213. tients with end-stage renal disease on hemodialysis. Kidney
311. Biasotto M, Chiandussi S, Zacchigna S, et al. A novel animal Int. 2004;65(5):1943–1946.
model to study non-spontaneous bisphosphonates osteonecro- 327. Aono Y, Yamazaki Y, Yasutake J, et al. Therapeutic effects
sis of jaw. J Oral Pathol Med. 2010;39(5):390–396. of anti-FGF23 antibodies in hypophosphatemic rickets/os-
312. Huja SS, Fernandez SA, Phillips C, Li Y. Zoledronic acid teomalacia. J Bone Miner Res. 2009;24(11):1879 –1888.
decreases bone formation without causing osteocyte death 328. Mirza MA, Hansen T, Johansson L, et al. Relationship
in mice. Arch Oral Biol. 2009;54(9):851– 856. between circulating FGF23 and total body atherosclerosis
313. Schulz A, Bressel M, Delling G. Activity of osteoclastic in the community. Nephrol Dial Transplant. 2009;24(10):
bone resorption in primary hyperparathyroidism—a com- 3125–3131.
parative electron microscopic and histomorphometric 329. Mirza MA, Larsson A, Melhus H, Lind L, Larsson TE.
study. Calcif Tissue Res. 1977;22 Suppl:307–310. Serum intact FGF23 associate with left ventricular mass,
314. Padagas J, Colloton M, Shalhoub V, et al. The receptor hypertrophy and geometry in an elderly population. Ath-
activator of nuclear factor-␬B ligand inhibitor osteopro- erosclerosis. 2009;207(2):546 –551.
tegerin is a bone-protective agent in a rat model of chronic 330. Hsu HJ, Wu MS. Fibroblast growth factor 23: a possible
renal insufficiency and hyperparathyroidism. Calcif Tissue cause of left ventricular hypertrophy in hemodialysis pa-
Int. 2006;78(1):35– 44. tients. Am J Med Sci. 2009;337(2):116 –122.
315. Silvestrini G, Ballanti P, Leopizzi M, et al. Effects of inter- 331. Dalal M, Sun K, Cappola AR, et al. Relationship of serum
mittent parathyroid hormone (PTH) administration on fibroblast growth factor 23 with cardiovascular disease in
SOST mRNA and protein in rat bone. J Mol Histol. 2007; older community-dwelling women. Eur J Endocrinol.
38(4):261–269. 2011;165(5):797– 803.
316. Kramer I, Loots GG, Studer A, Keller H, Kneissel M. Para- 332. Sylvester FA, Wyzga N, Hyams JS, et al. Natural history of
690 Dallas et al Osteocytes as Endocrine Cells Endocrine Reviews, October 2013, 34(5):658 – 690

bone metabolism and bone mineral density in children with formation in patients with quiescent Crohn’s disease. Gas-
inflammatory bowel disease. Inflamm Bowel Dis. 2007; troenterology. 2011;140(1):116 –123.
13(1):42–50. 335. Giuliani N, Ferretti M, Bolzoni M, et al. Increased osteo-
333. Pappa HM, Saslowsky TM, Filip-Dhima R, et al. Efficacy cyte death in multiple myeloma patients: role in myeloma-
induced osteoclast formation. Leukemia. 2012;26(6):
and harms of nasal calcitonin in improving bone density in
1391–1401.
young patients with inflammatory bowel disease: a ran-
336. Terpos E, Christoulas D, Katodritou E, et al. Elevated cir-
domized, placebo-controlled, double-blind trial. Am J culating sclerostin correlates with advanced disease fea-
Gastroenterol. 2011;106(8):1527–1543. tures and abnormal bone remodeling in symptomatic my-
334. Oostlander AE, Bravenboer N, Sohl E, et al. Histomor- eloma; reduction post-bortezomib monotherapy. Int J
phometric analysis reveals reduced bone mass and bone Cancer. 2012;131(6):1466 –1471.

Downloaded from https://academic.oup.com/edrv/article-abstract/34/5/658/2354663 by guest on 15 May 2020

Save the Date for Clinical Endocrinology Update (CEU) ,


September 26–28, 2013, Hyatt Regency New Orleans New Orleans, LA
www.endo-society.org/CEU2013

Das könnte Ihnen auch gefallen